153
Suppression of Collagen Induced Arthritis by NA-2 and Rutin through Regulation of RANKL Pathway A DISSERTATION SUBMITTED FOR THE PARTIAL FULFILMENT FOR THE DEGREE OF DOCTOR OF PHILOSOPHY BY ANUM GUL 2017 Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan

Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

Suppression of Collagen Induced Arthritis by NA-2

and Rutin through Regulation of RANKL Pathway

A DISSERTATION SUBMITTED FOR THE PARTIAL FULFILMENT FOR

THE DEGREE OF DOCTOR OF PHILOSOPHY

BY

ANUM GUL

2017

Dr. Panjwani Center for Molecular Medicine and Drug Research,

International Center for Chemical and Biological Sciences,

University of Karachi, Karachi-75270, Pakistan

Page 2: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

CERTIFICATE

It is certified that the thesis entitled, “Suppression of Collagen Induced Arthritis by NA-

2 and Rutin through Regulation of RANKL Pathway”, submitted by Ms. Anum Gul,

fulfills the requirement for the award of Doctor of Philosophy in Molecular Medicine. To

the best of my knowledge, no part of the work has been submitted for another degree or

qualification in any other institute.

Dr. Shabana U. Simjee

Research Supervisor

Page 3: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

Dedicated to

My Beloved Mother, Father

&

Siblings

Page 4: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

CONTENTS

Page. No

Acknowledgements...................................................................................................................... I

List of Tables ............................................................................................................................... II

List of Figures ........................................................................................................................... III

List of Abbreviation ................................................................................................................... V

Summary ................................................................................................................................... IX

صہ ال X ............................................................................................................................................ خ

CHAPTER 1

INTRODUCTION ....................................................................................................................... 1

1.1. Rheumatoid Arthritis and Its Prevalence ........................................................................ 2

1.2. Clinical Manifestation ......................................................................................................... 2

1.3. Risk factors and Pathogenesis of Rheumatoid Arthritis ............................................... 3

1.4. Reactive Oxygen and Nitrogen Species (ROS & RNS) .................................................. 5

1.4.1. Nitric Oxide ................................................................................................................. 7

1.4.2. Hydrogen Peroxide...................................................................................................... 8

1.5. Antioxidant Defense Mechanisms ..................................................................................... 8

1.5.1. Glutathione (GSH) .................................................................................................. 9

1.6. Cytokines in the Pathogenesis of Rheumatoid Arthritis................................................ 9

1.6.1. Pro-Inflammatory Cytokine: TNF-α ........................................................................ 10

1.6.2. Pro-inflammatory Cytokine: IL-1β .......................................................................... 11

1.7. RANK-RANKL-OPG in Bone Diseases ......................................................................... 11

1.7.1. RANK/RANKL Pathway in Rheumatoid Arthritis ................................................ 12

Page 5: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

1.7.2. RANK/RANKL Pathway and the Immune Response ............................................ 13

1.8. Overview of Treatment Strategies................................................................................... 14

1.8.1. Non-Biological Therapies for Rheumatoid Arthritis .............................................. 16

1.8.1.1. Non-Steroidal Anti-Inflammatory Drugs (NSAIDS)....................................... 16

1.8.1.2. Glucocorticoids (GCs) ....................................................................................... 17

1.8.1.3. Conventional DMARDs .................................................................................... 17

1.8.2. Biologics in the Treatment of RA ............................................................................ 19

1.8.2.1. Overview of Current Biologic Therapies ......................................................... 19

1.8.3. Non-Pharmacological Treatments for RA ............................................................... 20

1.9. Collagen Induced Arthritis (CIA) Rat Model ............................................................... 22

1.10. Compounds Used in the Current Study ....................................................................... 23

1.10.1. N-(2-Hydroxyphenyl) Acetamide (NA-2) ............................................................. 23

1.10.2. Rutin ......................................................................................................................... 24

1.10.3. Rutin Gold Nanoparticles (Rutin-GNPs) ............................................................... 24

1.11. Objectives .......................................................................................................................... 26

CHAPTER 2

METHODOLOGY.................................................................................................................... 27

2.1. Animal Care ........................................................................................................................ 28

2.2. Chemicals ............................................................................................................................ 28

2.3. Collagen Suspension Preparation.................................................................................... 28

2.4. Induction of Arthritis ........................................................................................................ 28

2.5. Treatment Strategy ............................................................................................................ 28

2.6. Clinical Assessment of Arthritis ...................................................................................... 30

2.6.1. Arthritic Score ........................................................................................................... 30

2.6.2. Hind Paw Volume Measurement ............................................................................. 30

Page 6: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

2.6.3. Body Weight Measurement ...................................................................................... 30

2.7. Histopathological Examination........................................................................................ 32

2.7.1. Fixation ...................................................................................................................... 32

2.7.2. Decalcification........................................................................................................... 32

2.7.3. Processing .................................................................................................................. 32

2.7.3.1. Dehydration ........................................................................................................ 32

2.7.3.2. Embedding .......................................................................................................... 32

2.7.3.3. Microtomy .......................................................................................................... 33

2.7.3.4. Staining ............................................................................................................... 33

2.8. Inflammatory Markers Analysis ..................................................................................... 33

2.8.1. Reactive Oxygen Species (ROS) and Antioxidant Measurement .......................... 33

2.8.1.1 Nitric Oxide Determination (NO) ...................................................................... 33

2.8.1.2. Peroxide Determination (PO) ............................................................................ 34

2.8.1.3. Glutathione Quantification (GSH) .................................................................... 34

2.9. Pro-Inflammatory Cytokines IL-1β and TNF-α Analysis ........................................... 35

2.9.1. IL-1β Measurement ................................................................................................... 35

2.9.2. TNF-α Measurement ................................................................................................. 35

2.10. Real time Reverse Transcriptase Polymerase Chain Reaction (Real Time RT-

PCR) ............................................................................................................................................ 36

2.10.1. RNA Isolation and Quantification ......................................................................... 36

2.10.2 The cDNA Synthesis................................................................................................ 36

2.10.3. Real time Reverse Transcriptase-PCR Amplification of cDNA .......................... 37

2.11. Immunohistochemistry for c-Fos, iNOS and pAkt in Spleen Tissue ....................... 37

2.11.1. Image Analysis ........................................................................................................ 41

2.12. Statistical Analysis ........................................................................................................... 41

Page 7: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

CHAPTER 3

RESULTS ................................................................................................................................... 43

3.1. Clinical Parameters of Collagen Induced Arthritis...................................................... 44

3.1.1. Effect of NA-2 on Clinical Parameters .................................................................... 44

3.1.1.1. Arthritic Score ................................................................................................... 44

3.1.1.2. Body Weight ...................................................................................................... 45

3.1.1.3. Paw Oedema ....................................................................................................... 45

3.1.2. Effect of Rutin on Clinical Parameters .................................................................... 48

3.1.2.1. Arthritic Score .................................................................................................... 48

3.1.2.2. Body Weight ...................................................................................................... 48

3.1.2.3. Paw Oedema ...................................................................................................... 50

3.1.3. Effect of Rutin-GNPs on Clinical Parameters of RA ............................................. 52

3.1.3.1. Arthritic Score .................................................................................................. 52

3.1.3.2. Body Weight ...................................................................................................... 52

3.1.3.3. Paw Oedema ...................................................................................................... 52

3.2. Histological Analysis of Knee Joints ............................................................................... 56

3.2.1. NA-2 Treatment ........................................................................................................ 56

3.2.2. Rutin Treatment......................................................................................................... 56

3.2.3. Rutin-GNPs Treatment ............................................................................................. 56

3.3. Measurement of Oxidative Stress Markers in Serum .................................................. 61

3.3.1. Effect of NA-2 Treatment ......................................................................................... 61

3.3.1.1. Serum Nitric Oxide (NO) .................................................................................. 61

3.3.1.2. Serum Peroxide (PO) ......................................................................................... 61

3.3.1.3. Glutathione (GSH) ............................................................................................. 61

3.3.2. Effect of Rutin on Oxidative Stress Parameters ...................................................... 63

Page 8: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

3.3.2.1. Serum Nitric Oxide (NO) ................................................................................. 63

3.3.2.2. Serum Peroxide (PO) ........................................................................................ 63

3.3.2.3. Serum Glutathione (GSH) ................................................................................. 63

3.3.3. Effect of Rutin-GNPs on Oxidative Stress Parameters .......................................... 64

3.3.3.1. Serum Nitric Oxide (NO) .................................................................................. 64

3.3.3.2. Serum Peroxide (PO) ......................................................................................... 64

3.3.3.3. Effect of Rutin-GNPs on Serum Glutathione (GSH) ....................................... 64

3.4. Measurement of Pro-Inflammatory Cytokines in Serum............................................ 67

3.4.1. Effect of NA-2 on Serum IL-1β and TNF-α Concentration ................................... 67

3.4.2. Effect of Rutin on IL-1β and TNF-α Concentration ............................................... 67

3.4.3. Effect of Rutin-GNPs on IL-1β and TNF-α Concentration .................................... 70

3.5. Gene Expression Studies Using Quantitative Real time RT-PCR ............................. 70

3.5.1. Effect of NA-2 Treatment ......................................................................................... 70

3.5.2. Effect of Rutin Treatment ......................................................................................... 72

3.5.3. Effect of Rutin-GNPs Treatment.............................................................................. 73

3.6. Immunohistochemistry of c-Fos, pAkt and iNOS in Spleen Tissue ........................... 76

3.6.1. Effect of NA-2 on c-Fos, pAkt and iNOS Protein Expression ............................... 76

3.6.2. Effect of Rutin on c-Fos, pAkt, and iNOS Protein Expression .............................. 81

3.6.3. Effect of Rutin-GNPs on c-Fos, pAkt, and iNOS Protein Expression................... 86

CHAPTER 4

DISCUSSION............................................................................................................................. 91

CONCLUSION ........................................................................................................................ 100

FUTURE IMPLICATIONS .................................................................................................. 101

REFERENCES ........................................................................................................................ 102

PERSONAL INTRODUCTION ........................................................................................... 126

Page 9: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

GLOSSARY ............................................................................................................................. 127

APPENDIX I ............................................................................................................................ 130

APPENDIX II .......................................................................................................................... 133

LIST OF PUBLICATIONS ……………………………………......................................134

Page 10: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

I

Acknowledgements

First, I am thankful to Almighty Allah, who has bestowed upon me His blessings that I was

able to complete my research successfully.

My heartfelt gratitude to my supervisor Dr. Shabana U. Simjee whose constant support,

expertise, and encouragement has made me able to contribute in the field of Molecular

Medicine. Without her devoted supervision, I could never finish this work. I am also grateful

to my former supervisor Dr. Talat Makhmoor. I cannot forget her initial training my whole

life. She always believed in me, and stood with me in my hard times.

I am very much thankful to Ms. Nadira Panjwani (Chairperson, Dr. Panjwani Memorial Trust)

who has established this international research center and provided us the opportunities for

research of international level and showing our strengths to the rest of the world. Moreover, I

am indebted to Prof. Dr. Atta-ur-Rahman F.R.S., N.I., H.I., S.I., T.I. (Patron in Chief) and Prof. Dr. M.

Iqbal Choudhary H.I., S.I., T.I. (Director, PCMD) for running such a remarkable institution and

for providing us the best working environment.

I am very thankful to Prof. Dr. Shaheen Faizi S.I., Dr. Raza Shah T.I., all my teachers, and PCMD

faculty members.

I am grateful to all my friends who provided me support in hard times, especially Areeba

Anwar and Maryam Mazhar.

I am very much thankful to my loving mother Saeeda Haider, father Ghulam Haider and

siblings Meisha Gul and Ali Shayan Haider for their unconditional support throughout my

studies.

Page 11: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

II

List of Tables

Table No. Title Page No.

1.1 Approved biologic therapies of rheumatoid arthritis………………...21

2.1 Treatment regime used for the testing of compounds

for anti-arthritic activity in collagen induced arthritis

model in rats…………………………………………………………29

2.2 Scoring system used to assess severity of paw

inflammation………………………………………………………...31

2.3 Composition of RNA/Primer mixture……………… ……................38

2.4 Composition of cDNA synthesis mixture…………………...............38

2.5 Real time RT- PCR reaction mixture recipe………...........................39

2.6 Steps involved in real time RT-PCR reaction……………..............39

2.7 Primer sequences, annealing temperature and expected

product size……………………………………….............................40

2.8 List of antibodies for immunohistochemistry……………….............42

3.1 Histological scoring system adopted to monitor the histological

changes in arthritic and non-arthritic rats…………………………...57

Page 12: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

III

List of Figures

Figure 1.1. Factors involved in the progression of rheumatoid arthritis .............................. 4

Figure 3.1. Effect of NA-2 on mean arthritic severity score in rats induced with collagen

induced arthritis .................................................................................................. 46

Figure 3.2. Effect of NA-2 on mean change in body weight .............................................. 46

Figure 3.3. Effect of NA-2 on mean change in hind paw volume after arthritis induction.

...................................................................................................................................................... 47

Figure 3.4. Effect of rutin on mean arthritic score ............................................................... 49

Figure 3.5. Mean change in body weight of arthritic and non-arthritic animals................ 49

Figure 3.6. Mean change in paw volume following treatment with rutin .......................... 51

Figure 3.7. Effect of rutin-GNPs on arthritic score.. ........................................................... 53

Figure 3.8. Effect of rutin-GNPs on mean change in body weight..................................... 54

Figure 3.9. The mean change in paw volumes following treatment with rutin-GNPs ...... 55

Figure 3.10. Effect of NA-2 treatment on joint destruction in CIA rats………………….58

Figure 3.11. Effect of rutin treatment on Knee joints sections from CIA rats……………59

Figure 3.12. Effect of rutin-GNPs treatment on Knee joints sections from CIA rats…….60

Figure 3.13. Effect of NA-2 on serum NO, PO and GSH level in arthritic and non-arthritic

rats ....................................................................................................................... 62

Figure 3.14. The NO, PO and GSH concentration measured following rutin treatment..... 65

Figure 3.15. Serum NO, PO and GSH concentration measured after treatment of rutin-

GNPs.. ................................................................................................................. 66

Figure 3.16. Effect of NA-2 on IL-β and TNF-α concentration.. ......................................... 68

Figure 3.17. Effect of rutin on IL1-β and TNF- α concentration .......................................... 69

Figure No. Title Page No.

Page 13: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

IV

Figure 3.18. Effect of rutin-GNPs on IL-1β and TNF-α concentration ................................. 71

Figure 3.19. The effect of NA-2 on fold change in RANK, RANKL, NF-κB, c-fos, c-jun

and Akt gene expression .................................................................................... 74

Figure 3.20. Effect of rutin on RANK, RANKL, NF-κB, c-fos, c-jun and Akt gene

expression............................................................................................................ 74

Figure 3.21. Expression of RANK, RANKL, NF-κB, c-fos, c-jun and Akt genes following

rutin-GNPs treatment. ........................................................................................ 75

Figure 3.22. Effect of NA-2 on c-Fos, pAkt and iNOS Protein Expression ........................ 77

Figure 3.23. Effect of NA-2 on immunohistochemistry of c-Fos in spleen tissue. ............. 78

Figure 3.24. Effect of NA-2 on Immunohistochemical analysis of pAkt in arthritic and

non-arthritic rats.. ............................................................................................... 79

Figure 3.25. Immunohistochemical analysis of iNOS expression after treatment with NA-2

............................................................................................................................. 80

Figure 3.26. Effect of rutin on c-Fos, pAkt and iNOS protein expression. .......................... 82

Figure 3.27. Immunohistochemistry of c-Fos in rutin treated animals…………………...83

Figure 3.28. Immunohistochemical analysis of pAkt in rutin treated group……………..84

Figure 3.29. Photomicrograph of iNOS expression in rutin treated animals……………..85

Figure 3.30. Effect of rutin-GNPs on c-Fos, pAkt and iNOS expression………………...87

Figure 3.31. Effect of rutin-GNPs on Immunohistochemical analysis of c-Fos .................. 88

Figure 3.32. Immunohistochemistry of pAkt on rutin-GNPs treated rats ............................ 89

Figure 3.33. Immunohistochemistry of iNOS in spleen tissue of rutin-GNPs treated group

...................................................................................................................................................... 90

Page 14: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

V

List of Abbreviation

ACPA Anti-citrullinated protein antibody

AIA Adjuvant-induced arthritis

ANOVA Analysis of variance

AP1 Activator protein 1

ASA Acetylsalicylic acid

ATF Activating transcription factor 3

CAT Catalase

CD Cluster of differentiation

cDNA Complementary DNA

CFA Complete Freund’s adjuvant

CIA Collagen induced arthritis

COX Cyclo-oxygenase

CP Ceruloplasmin

CSF1R Colony Stimulating Factor 1 Receptor

c-Src Proto-oncogene tyrosine-protein kinase Src

CTGF Connective tissue growth factor

CTLA Cytotoxic T lymphocyte antigen

CyA Cyclosporine A

DMARDs Disease modifying anti-rheumatic drugs

DNTB 5, 5’-dithiobis 2-nitrobenzoic acid

EBV Epstein-Barr virus

Page 15: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

VI

eNOS Endothelial nitric oxide synthase

ERK Extracellular signal regulated kinase

Fab Fragment of antigen binding

Fc Fragment of crystallization

GAB2 GRB2 Associated Binding Protein 2

GAPDH Glyceraldehyde 3-phosphate dehydrogenase

GC Glucocorticoid

GNPs Gold nanoparticles

GSH-Px Glutathione peroxidase

GST Glutathione S-transferase

H & E Hematoxylin and eosin

H2O2 Hydrogen peroxide

HCQ Hydroxychloroquine

HLA Human leukocyte antigen

HLA-DRB1 Human leukocyte antigen- DR beta 1

IBD Inflammatory bowel disease

IFA Incomplete Freund’s adjuvant

IFN-γ Interferon gamma

IKK2 Inhibitor of nuclear factor-kappaB kinase subunit beta

IL-1β Interlukin-1-beta

iNOS Inducible NOS

JNK c-Jun N-terminal kinase

LPS Lipopolysaccharide

Page 16: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

VII

MAF Macrophage activating factor

MAPK Mitogen activated protein kinase

M-CSF Macrophage colony-stimulating factor

MK2 MAP Kinase-Activated Protein Kinase 2

MMPs Matrix metalloproteinases

MTX Methotrexate

NA-2 N-(2-hydroxyl phenyl) acetamide

NF-κB Nuclear factor-κB

nNOS Neuronal NOS

NO Nitric oxide

NOSs NO synthases

NSAIDs Non-steroidal anti-inflammatory drugs

OPG Osteoprotegerin

p38 Protein 38

PAD Peptidylarginine deiminase

PBS Phosphate buffered saline

PCR Polymerase Chain Reaction

PGE2 Prostaglandin E2

PI3K Phosphotidyl-inositol-3- phosphate

PsA Psoriatic arthritis

RA Rheumatoid arthritis

RANK Receptor activator of NF-κB

RANKL Receptor activator of NF-κB ligand

Page 17: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

VIII

RF Rheumatoid Factor

RNS Reactive nitrogen species

ROS Reactive oxygen species

S.E.M Standard error of mean

SAARDs Slow acting anti-rheumatic drugs

SE Shared epitope

SOD Superoxide dismutase

SPSS Statistical package for social sciences

SSZ Sulfasalazine

STAT3 Signal transducer and activator of transcription 3

TMB 3,3',5,5'-Tetramethylbenzidine

TNFR Tumor necrosis factor receptor

TNF-α Tumor necrosis factor alpha

TRAF TNF receptor associated factor

Treg cells Regulatory T cells

Page 18: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

IX

Summary

Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease of unknown

etiology that results in inflammation in the synovial joint, and irreversible bone and cartilage

destruction. It includes a complex series of events involving environmental and genetic factors

that leads to the pathogenesis of RA. Various drugs have been used for RA treatment but none

is able to prevent the bone destruction that is the most severe outcome of RA progression.

RANK and RANKL are involved in the maintenance of normal bone physiology and any

alteration in the level of these molecules is responsible for bone pathologies. In osteoclast

research, RANKL is becoming an important therapeutic target since it binds to mature

osteoclasts and causes induction of bone resorbing activity. Beside this, RANKL and RANK

are also key players of the inflammatory cascade in human and animal models of arthritis. In

the current study, we explored the effect of N-(2-hydroxyl phenyl) acetamide (NA-2), rutin,

indomethacin + rutin, and rutin-GNPs on the attenuation of RANK, RANKL, and other pro-

inflammatory mediators downstream of RANKL pathway to understand the possible

mechanism behind the anti-arthritic effect of these compounds in collagen induced arthritis

(CIA) model in rats. The progression of arthritis was analyzed by clinical parameters such as,

body weight, paw volume and arthritic score that showed marked improvement after treatment

with the compounds. Similarly, the level of inflammatory markers in serum such as, TNF-α,

IL-1β, NO and PO after compounds treatment were also reduced with parallel increase in the

GSH level. The gene expression analysis of RANK, RANKL, c-fos, c-jun, Akt, and NF-κB

was done in spleen tissue by real time PCR and the compounds treatment successfully

downregulated the expressions of these genes. Immunohistochemical analysis of iNOS, c-Fos,

and pAkt in spleen tissue also showed marked suppression in the protein expression in arthritic

rats, treated with the above cited compounds. These findings suggest that NA-2, rutin (both

as a single agent and in combination with indomethacin or gold nanoparticles) are potent anti-

rheumatic agents and can inhibit the inflammatory response and destruction of bone and

cartilage possibly by modulation of RANK/RANKL pathway.

Page 19: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

X

صہ ال خ

ڈیر وائ ٹس ا وماٹ ھرائ م کیآرت ے۔ اس مرض م یدائ سم م ںیمرض ہ اعث ج ے ب لوم وجہ ک امع ںین

ہ نے ٹھوں ک یاپ عمال جوڑوں اور پ ت س ا ا عت ک وت مداف الف ق س یخ ل واپ اب اق سوزش اور ن ڈ ی وںیہ

مزور یک ہ یک با ے۔ یاور ت ا ہ وت اعث ہ ا ب ہیک لوں ک دہیچیمرض پ لہ وار مرح س ل س صورت م ی ںی

ے اور اس عمل م ا ہ وت ر ہ صہ ل یاتیاور ماحول یاتینیج ںیظاہ ےیعوامل ح ڈی۔ رںیہ ت وائ وماٹ

شمار ادو ے الف ب ے خ ٹس ک ھرائ وت اتیآرت عمال ہ ت س ہ یا وئ ںیہ یآر گر ک ھ یم ڈ یب یک وںیدوا ہ

مزور ہ یک ے، دور ن لو ہ ہ اک پ سے خطرن سب ا ہ اس مرض ک سک ںیجو ک ر کی۔ ریک کیاور ر ن ن

سےیدو ا لیا زیکیمال ہ ول ڈ ںی ز یک وںیجو ہ ارمل ف وجین ہ یال تے ھ رار رک رق و ب اور ان ںیک

عمول ںیم بد یم ڈ یلیت نم د وںیماریب یک وںیہ و ج کی۔ رںیہ یتیک ڈ لیا ن ے وںیہ ے امراض ک ک

الج م ڈ کیا ںیع ے جو ہ دف ہ م ہ ے خل اںیاہ ے وال رن مزور ک سٹ وںیک سٹ،یاو ال ھ مل وک سات ے ک

ر ھڈ الوہ ر اںیک ے ع ے۔ اس ک ا ہ رت مزور ک کیک کیاور ر ن وں م لیا ن سان ے ںیان وروں ک اور جان

ٹس ماڈل م ھرائ سوزش پ ںیآرت ے م دای رن ہ ںیک ے رت ردار ادا ک م ک ئے حالںیاہ ہی۔ اس ل حق قیت

و، ر نیا ںیم ن،یاے ٹ نیر وٹ ڈوم وٹ ھ ان سات ے سنیک ھا نیاور ر ت ڈ ن وٹ ول ویگ ارٹ ن لزیپ ک

ول و ک چا گ جنیک الف جان ے خ ٹس ک ھرائ ے آرت ے وال ون سے ہ و ر ای بات ک ہ ان مرک اک ک،یت ن

کیر کیاور ر لیا ن ھوے م لیا ن ات سوزش پ دیمز ںیپ ے مال دای ے وال رن زیکیک ے ول ٹان ھ ر گ پ

لوم ک و مع نہ عمل ک ک ر اور مم ے اث و طب ایک نے ک ڑھ ے ب ے درد ک کے۔ جوڑوں ک س یجا

رزیرامیپ سایج ٹ ٹھ نجا حجم اور گ ے وزن، پ سم ک ہ ج کور ک ایک س لوم ک یا سے مع ایگ ایمدد

ست ے ا بات ک عد ان پ عمالاور ان مرک ے ب رزیرامیک تر ںیم ٹ ہ ھید یب ئ یک طرح ان ی۔ اسیگ

ے س وں ک ٹس زدہ چوہ ھرائ سے آرت عمال ت س ے ا بات ک وں م رمیمرک مون سوزش پ ںین ے دای رن ک

ے مال زیکیوال سایج ول ہ آئ ا،یون ب لیا یک فا،یا فیا نیا یٹ ٹ سائ ل ٹرک آک ائ ر ڈین اور پ

سائ ے م یک ڈیآک ٹان ھ و گ سطحوں ک ھا ںی ات لوٹ ے م یون کیاور گ ڑھان و ب سطح ک ام ںی ابیک

ے۔ ج وئ ت ہ اب ر ر یاتینیث سطح پ ک،ی کیر ن وس، س یس ل،یا ن ے ٹ یف نیاور ا ،یجون، اے ک

ا ب فیا اپ ل یک ہ ت ھے، ر یجو ک ئے ت ئے گ صل ک سے حا لی م پ ئ ائ سے یآر ک یس یٹ مدد

ے م ے جان ر ل سطح پ م و ک بات ان ک ئے اور مرک چے گ ام ںیجان ے۔ ام ابیک ٹوکیرہ س وہ لیمیون ک

سٹیٹ ل سے ت وس، س یآئ ںیم ی وس اور پ ین ے ٹ یف روٹ یاے ک زیپ ا ن وا پ ٹا ہ ھ و گ ۔ ان ایک

سے ج تائ ہین ہ ا ے ک ا ہ وت ر ہ و نیظاہ ن،یر اے ٹ نیر وٹ ڈوم وٹ ھ ان سات ے سنیک ھا نیاور ر ت وٹ

ڈ ن ول ویگ ارٹ ن لزیپ و ک الف ق ے خ ٹس ک ھرائ ہ یآرت تے ھ ر رک ک،یاور ر ںیاث کیر ن اور لیا ن

سے پ ردہ د دایان سرےک سوزش و ڈ ی سوزش، ہ ر و ک داز ہ ر ان ر اث ل وںیعوامل پ ارٹ یک جیاور ک

ہ با ہ یک یت ے رت ھام ک ۔ںیروک ت

Page 20: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

1

CHAPTER 1

INTRODUCTION

Page 21: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

2

1.1. Rheumatoid Arthritis and Its Prevalence

Rheumatoid arthritis (RA) is an autoimmune disorder that leads to disability, bone and

cartilage damage ultimately leading to pulmonary and cardiovascular disorders (Picerno,

Ferro, Adinolfi, Valentini, Tani, and Alunno, 2015). The prevalence of RA is 0.5–1% in

Western adult populations and is comparatively similar across Asia, South Africa, North

America and Europe (Jacobsson, Hanson, Knowler, Pillemer, Pettitt, Mccance, and Bennett,

1994). Annually, the incidence of RA is between 0.15 and 0.88 per 1000 and females are

affected two to three times more as compared to male population. The disease can occur at

any stage of life but individuals with 45 to 65 years of age are more prone to develop this

condition (Scott and Wolfe, 2010).

1.2. Clinical Manifestation

The symptoms of RA progression vary but some features are same in all individuals like

morning joint stiffness, pain and movement impairment. Typically, the symptoms include

generalized inflammation, the individual experience weight loss, fatigue, low grade fever and

exhaustion. The most commonly affected area in the body as a result of arthritis progression

are joints especially small joints of hand and feet along with some larger joints and their

inflammation leads to swelling of the synovium.

In RA, the damage to articular cartilage results in anatomical changes in joints over the course

of time. The blend of these constant changes prompts some clinically recognized patterns of

deformation, for example, swan-neck and boutonnière disfigurements of the fingers, hammer-

toe distortions in the feet, the ulnar deviation of the hands, and others. RA patients with severe

joint disease develop extra articular complications like lung and cardiac involvement,

vasculitis and in some RA patients the increase in mortality rate is because of high occurrence

of cardiovascular disease (Wållberg-Jonsson, Johansson, Ohman, and Rantapää-Dahlqvist,

1999; Watson, Rhodes, and Guess, 2003).

Page 22: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

3

1.3. Risk factors and Pathogenesis of Rheumatoid Arthritis

Despite intensive research, the pathogenesis of RA is poorly understood and a complex

interaction among genetic and environmental factors and continuous activation of both innate

and adaptive immunity results in breakdown of immunological tolerance, antigen specific B

and T cell activation and abnormal presentation of autoantigens. All these events eventually

lead to bone destruction and synovial hyperplasia resulting in deformity and swelling of joint

and ultimately systemic inflammation (Fig. 1.1) (MacGregor, Snieder, Rigby, Koskenvuo,

Kaprio, Aho, and Silman, 2000; McInnes and Schett, 2011; Picerno, Ferro, Adinolfi,

Valentini, Tani, and Alunno, 2015).

The most common genetic risk factor is associated with human leukocyte antigen (HLA)

DRB1 locus that contain a conserved sequence of amino acid QKRAA called the shared

epitope (SE), which is associated with RA susceptibility (Gregersen, Silver, and Winchester,

1987). On the other side, environmental factors such as smoking, infectious agents and

gastrointestinal microbiome also participates in the RA pathogenesis. The interactions

between HLA-DRB1 alleles and smoking synergistically increase the risk of RA (Klareskog,

Rönnelid, Lundberg, Padyukov, and Alfredsson, 2008; Scher and Abramson, 2011).

Mechanistically, smoking causes the induction of peptidylarginine deiminase (PAD) enzymes

that cause post translational modification of protein resulting in citrullination of protein that

elicits anti-citrullinated protein antibody (ACPA) response against citrullinated proteins

(Vincent, De Keyser, Masson-Bessière, Sebbag, Veys, and Serre, 1999; De Rycke, Peene,

Hoffman, Kruithof, Union, Meheus, Lebeer, Wyns, Vincent, and Mielants, 2004). Infectious

agents such as E. coli, cytomegalovirus, Epstein-Barr virus (EBV) and proteus species and

their products (e.g. heat shock proteins) have been suspected as potential triggers of RA but

the mechanisms of pathogenesis are not known completely. It is postulated that some form of

molecular mimicry might be involved (Auger and Roudier, 1997; Kamphuis, Kuis, de Jager,

Teklenburg, Massa, Gordon, Boerhof, Rijkers, Uiterwaal, and Otten, 2005). In RA

pathogenesis, pro-inflammatory cytokines and chemokines produced by synovial cells are

also key players (Kirkham, Lassere, Edmonds, Juhasz, Bird, Lee, Shnier, and Portek, 2006).

In addition to this, macrophages, the innate effector cells, with antigen presentation and

phagocytic ability also serve as central effectors of synovitis. The infiltration and activation

Page 23: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

4

Figure 1.1. Factors involved in the progression of rheumatoid arthritis. Environmental

and genetic factors lead to breakdown of tolerance to self-proteins having citrulline residues

generated by post-translational modification. In bone marrow or secondary lymphoid tissues,

this anti-citrulline protein response is generated in B and T cell. After these events,

inflammatory response is observed in the joints that may involve neurologic, microvascular,

biomechanical and other pathways. Synovitis occurs as a result of positive feedback loops that

promote systemic disorders that ultimately lead to RA. RF denotes rheumatoid factor and

ACPA denotes anti–citrullinated protein antibody (adapted from McInnes and Schett, 2011).

Page 24: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

5

of macrophages and T cells in the synovium results in the production of various growth factors

and cytokines like TNF-α (tumor necrosis factor-α), TGF- β (transforming growth factor β),

interleukins i.e. IL-1, IL- 2, IL- 6, IL-8, IL-10, IL-17, insulin like growth factor and platelet

derived growth factors. All these effector molecules are involved in cartilage and bone

destruction, synovitis and systemic complications (Stolt, Bengtsson, Nordmark, Lindblad,

Lundberg, Klareskog, and Alfredsson, 2003; Bresnihan, and Tak, 2005; Haringman, Gerlag,

Zwinderman, Smeets, Kraan, Baeten, McInnes, Maruotti, Cantatore, Crivellato, Vacca, and

Ribatti, 2007; Cornish, Campbell, McKenzie, Chatfield, and Wicks, 2009). In the synovium,

neo-angiogenesis which is the growth of new capillary blood vessels is also observed in the

persistence of inflammation in patients of RA. The release of different enzymes and

metalloproteinases that are damaging to bone and cartilage are also released by the cells of

synovium and articular cartilage (Smith and Haynes, 2002).

B cells are also key players in the progression of RA. Synovium infiltration with B cells results

in its differentiation into plasma cells that produces Rheumatoid Factor (RF). In addition,

activation of synovial fibroblast leads to the destruction of matrix tissues by the action of

enzymes like collagenases and metalloproteinases. The overall result of all these activities is

cartilage and bone erosion, pannus formation and joint swelling with destruction of

periarticular structures (Smith and Haynes, 2002; Stolt, Bengtsson, Nordmark, Lindblad,

Lundberg, Klareskog, and Alfredsson, 2003).

1.4. Reactive Oxygen and Nitrogen Species (ROS & RNS)

Reactive oxygen species (ROS) are important mediators in the cellular signaling cascades

(Irani, 2000). Appropriate levels of ROS are required for several physiological processes and

have beneficial effects such as in tissue regeneration, wound healing and protection from

pathogens (Bhattacharyya, Chattopadhyay, Mitra, and Crowe, 2014; Datta, Kundu, Ghosh,

De, Ghosh, and Chatterjee, 2014). Deleteriously higher levels of ROS lead to disease

development and progression especially neurodegenerative diseases, cardiovascular diseases,

cancer and chronic inflammation (Halliwell and Cross, 1994; Bhattacharyya, Chattopadhyay,

Mitra, and Crowe, 2014). Some of the factors which trigger increased production of ROS

include exposure to UV radiation, alcohol consumption, cigarette smoking, some drugs,

Page 25: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

6

inflammatory disorders and ischemia–reperfusion injury (Halliwell and Cross, 1994; Iuchi,

Akaike, Mitsui, Ohshima, Shintani, Azuma, and Matsumoto, 2003; Mallick, Yang, Winslet,

and Seifalian, 2004; Roessner, Kuester, Malfertheiner, and Schneider-Stock, 2008; Wu and

Cederbaum, 2009; Jia, Koonce, Griffin, Jackson, and Corry, 2010; Deavall, Martin, Horner,

and Roberts, 2012)

The family of ROS is comprised of peroxide, superoxide and hydroxyl radicals which are

involved in oxidation of proteins, nucleotides, polysaccharides, DNA and lipids by the

unpaired free radicals. In inflammation, ROS are produced that promotes release of variety of

potentially harmful enzymes, neutrophils degranulation and the destruction of bone and

cartilage (Stamp, Khalilova, Tarr, Senthilmohan, Turner, Haigh, Winyard, and Kettle, 2012).

In patients with RA several inflammatory products mediated by ROS have been identified in

the peripheral blood and synovial fluid (Hadjigogos, 2003; Filippin, Vercelino, Marroni, and

Xavier, 2008; Winyard, Ryan, Eggleton, Nissim, Taylor, Faro, Burkholz, Szabó-Taylor, Fox,

Viner, Haigh, Benjamin, Jones, Whiteman, 2011).

ROS and RNS are highly reactive entities which are very difficult to be determined directly

in vivo. However, measurement of resulting effects of ROS and RNS on various cellular and

extracellular components is more practical (Vasanthi, Nalini, and Rajasekhar, 2009). Still

there is no clear proof that whether ROS are involved in initiation of autoimmunity or

ameliorate chronic inflammation in RA. Under physiological conditions, T cell functions are

regulated by NO, but overproduction of NO may lead to functional changes in T lymphocytes

(Nagy, Koncz, Telarico, Fernandez, Érsek, Buzás, and Perl, 2010). Some researchers have

identified the relationship between the RA disease and the level of serum nitrite (Onur, Akıncı,

Akbıyık, and Ünsal, 2001; Choi, 2003).

There are various antioxidant systems present in cells required for defense against free radical

damage. Red blood cells in human circulation scavenge H2O2 and O2- that are produced by

activated neutrophils as a result of redox reaction utilizing superoxide dismutase (SOD),

catalase (CAT) and glutathione peroxidase (GSH-Px) dependent mechanisms. There is a

controversy regarding antioxidant status in erythrocytes and serum of RA patients, since few

studies have shown decreased while others have reported unaltered activity of antioxidant

Page 26: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

7

enzymes (Cimen, Çimen, Kacmaz, Öztürk, Yorgancioğlu, and Durak, 2000; Nivsarkar, 2000;

Afonso, Champy, Mitrovic, Collin, and Lomri, 2007). Oxidative stress mediates inflammatory

responses in the arthritic joint (Henrotin, Bruckner, and Pujol, 2003; Datta, Kundu, Ghosh,

De, Ghosh, and Chatterjee, 2014). Hydrolysis of collagen and degradation of the extracellular

matrix in cartilage are also attributed to the intra-articular ROS which activates

metalloproteinase resulting in the development of osteoarthritis. (Goldring, 2003; Henrotin,

Bruckner, and Pujol, 2003; Yamazaki, Fukuda, Matsukawa, Hara, Matsushita, Yamamoto,

Yoshida, Munakata, and Hamanishi, 2003; Yamazaki, Fukuda, Matsukawa, Hara, Yoshida,

Akagi, Munakata, and Hamanishi, 2003; Fay, Varoga, Wruck, Kurz, Goldring, and Pufe,

2006; Regan, Bowler, and Crapo, 2008; Yu, Yi, Yuh, jin Park, Kim, Bae, Ji, Kim, Park, and

Kim, 2012).

1.4.1. Nitric Oxide

Nitric oxide (NO) is a molecule with a very short half-life. In various physiological functions,

NO plays an important role such as mitochondrial functions, regulation of inflammation,

apoptosis and blood vessel tone (Brown, 1999; Beltrán, Mathur, Duchen, Erusalimsky, and

Moncada, 2000). It is synthesized from L-arginine by NO synthases (NOSs) such as neuronal

NOS (nNOS), inducible NOS (iNOS) and endothelial NOS (eNOS) (Chiavaroli, Giannini, De

Marco, Chiarelli, and Mohn, 2011). NO is produced in different cell types such as phagocytic

neutrophils, leukocytes, chondrocytes, and synoviocytes. NO is mainly involved in regulation

of immune system, neurotransmission, and vasodilatation (Labunskyy and Gladyshev, 2013).

Furchgott and Zawadzki, first identified NO as endothelium derived relaxant factor (Furchgott

and Zawadzki, 1980). They observed the importance of intact endothelium for acetyl choline-

induced blood vessel relaxation. This observation led to the discovery of endothelium-derived

relaxant factor, identified as NO (Palmer, Ferrige, and Moncada, 1987). In rodent

macrophages, LPS and inflammatory cytokines like IL-1-β, IFN-γ and TNF-α induces the

expression of iNOS in vitro that leads to long term production of NO in large amount (Brown,

1999). Several studies have reported higher levels of endogenous NO synthesis in patients

with RA, suggesting the stimulating role of NO in the progression of RA (Farrell, Blake,

Palmer, and Moncada, 1992; Pham, Rahman, Tobin, Khraishi, Hamilton, Alderdice, and

Page 27: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

8

Richardson, 2003). The regulation of iNOS expression occurs at transcriptional level whereas

intracellular Ca2+ regulates eNOS and nNOS. In the inflamed joint, different cells such as

macrophages, osteoblasts, fibroblasts, neutrophils, osteoclasts and endothelial cells produces

NO in the synovium of RA patients (Van'T Hof and Ralston, 2001; Nagy, Clark, Buzás,

Gorman, and Cope, 2007; Firestein, Budd, Gabriel, McInnes, and O'Dell, 2012). It has been

reported that NOS inhibition contribute to decrease in disease progression in experimental RA

(McCartney-Francis, Allen, Mizel, Albina, Xie, Nathan, and Wahl, 1993).

1.4.2. Hydrogen Peroxide

Hydrogen peroxide is a member of reactive oxygen species (ROS) family that are produced

as a result of oxidative metabolism (Bedard and Krause, 2007). H2O2 is also involved in

regulation of many cellular events including cell proliferation, transcription factors activation,

and apoptosis (Rojkind, Dominguez-Rosales, Nieto, and Greenwel, 2002). H2O2 modulate

many different signaling pathways including p38, ERK, MAPK, JNK and PI3K/Akt (Gabbita,

Robinson, Stewart, Floyd, and Hensley, 2000; Hensley, Robinson, Gabbita, Salsman, and

Floyd, 2000). H2O2 can stimulate the macrophages to release high mobility group 1 protein

resulting in augmentation of pro-inflammatory stimuli or modify expression of leukocyte

endothelial adhesion and leukocyte adhesion molecule (Tang, Shi, Kang, Li, Xiao, Wang, and

Xiao, 2007; Sen and Roy, 2008). Hydrogen peroxide and superoxide radical induces the

release of TNF-α and interleukins from T lymphocytes. It induces endothelial cells that results

in production of adhesive molecules, growth factors and inflammatory cytokines by immune

cells thereby aggravating inflammation and tissue destruction (Los, Dröge, Stricker, Baeuerle,

and Schulze‐ Osthoff, 1995; Feldmann, Brennan, and Maini, 1996).

1.5. Antioxidant Defense Mechanisms

There are multiple defense systems in our body collectively called antioxidants. Their function

is to fight against free radical-induced oxidative stress. In humans, these antioxidants are

present in erythrocytes, plasma as well as serum. Serum antioxidants include ascorbate,

transferrin and ceruloplasmin (CP). Erythrocytes have glutathione S-transferase (GST),

superoxide dismutases (SODs), glutathione peroxidase (GPx), glutathione (GSH) and catalase

(Cat) that work in co-ordination to handle intracellular oxidative stress (McCord and Edeas,

Page 28: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

9

2005; Valko, Leibfritz, Moncol, Cronin, Mazur, and Telser, 2007). Imbalance in oxidant to

antioxidant ratio in the body leads to potentially destructive effects on cellular functions and

activities (Soneja, Drews, and Malinski, 2005; Blair, 2006).

1.5.1. Glutathione (GSH)

Glutathione (L-γ-glutamyl-L-cisteinylglycine) is an important part of antioxidant defense

mechanism. In prokaryotes and eukaryotes, it is present in cells as a tripeptide, a non-protein

sulfhydryl molecule (Pastore, Federici, Bertini, and Piemonte, 2003). In normal metabolic

process hydrogen peroxides are produced, GSH play central role in the protection of cells

from harmful effects of peroxide (PO) (Dröge, Schulze-Osthoff, Mihm, Galter, Schenk, Eck,

Roth, and Gmünder, 1994). In the cell, the enzyme glutathione reductase converts the inactive

oxidized form of glutathione (GSSG) into active reduced form GSH. Glutathione reductase is

constitutively active but in oxidative stress, the level of GSH is high (Pastore, Federici,

Bertini, and Piemonte, 2003). Reduced levels of GSH may contribute to

inflammatory/immunomediated diseases, inflammation, cellular injury in RA, vascular injury

and tissue damage (Collard, Agah, and Stahl, 1998).

1.6. Cytokines in the Pathogenesis of Rheumatoid Arthritis

Cytokines are small proteins from diverse group, which includes chemokines, interferon,

interleukins, monokines, colony stimulating factors and lymphokines that are synthesized

during immune response. These cytokines bind to their respective receptors and perform their

function that is characterized by pleiotropy and redundancy. Cytokine receptors are either

heterodimeric or heterotrimeric grouped into families with sharing of one receptor subunit by

each member of the receptor family. Imbalance or uncontrolled production of these cytokines

leads to inflammatory response and may lead to autoimmunity. In case of rheumatoid arthritis,

the secretion of cytokines in synovium alters with the disease progression. In the early stage

of RA, Th cells in the synovium express IL-2, IL-4, IL-13, IL-15 and IL-17. Whereas in the

synovium of full blown RA, Th cells express IFN-γ, TNFα and IL-10. However, expression

of IL-2, IL-4, IL-5 and IL-13 cytokines by Th cells is either very low or completely absent.

The underlying mechanism of fluctuating cytokine profile with the progression of disease is

still not completely understood (Cope, 2008).

Page 29: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

10

1.6.1. Pro-Inflammatory Cytokine: TNF-α

TNF-α is one of the most important primary cytokines of the cascade involved during the

course of RA and regulates the secretion of other pro-inflammatory cytokines present in the

synovial tissue. TNF-α is largely produced by immune cells such as neutrophils, natural killer

cells, endothelial cells, activated lymphocytes and macrophages (Monaco, Nanchahal, Taylor,

and Feldmann, 2015). TNF-α stimulates synovial fibroblasts and subsequent production of

CTGF (connective tissue growth factor), that leads to the osteoclasts activation resulting in

the disturbances of cartilage homeostasis and ultimately destruction of the joint (Monaco,

Nanchahal, Taylor, and Feldmann, 2015). Furthermore, the functions of Treg cells are

inhibited by TNF-α by inducing resistance in effector T cells against Treg-mediated

suppression (Komatsu and Takayanagi, 2015).

TNF-α mediates clearly distinguished range of RA elements. Beginning the course of disease

through activation of osteoclasts and chondrocytes to cause articular destruction and bone

resorption thereafter, induction of endothelial cells and amplification of chemokines resulting

in pannus formation along with stimulation of PGE2 synthesis and nociceptors sensitization

to induce pain and fever (Brennan and McInnes, 2008). Since a number of TNF-α inhibitors

have shown a great deal of success for the treatment of RA, therefore TNF-α is a center of

attention for immunologists and rheumatologists. TNF-α inhibitors decrease inflammation

and offers bone protection in patients of RA (Scott and Kingsley, 2006). It has been found

that inhibition of TNF-α reduces bone resorption in postmenopausal osteoporosis in vivo

(Charatcharoenwitthaya, Khosla, Atkinson, McCready, and Riggs, 2007). TNF-α induces

bone destruction in a manner very similar to other inflammatory cytokines, including IL-1 and

M-CSF (Kudo, Fujikawa, Itonaga, Sabokbar, Torisu, and Athanasou, 2002; Kitaura, Zhou,

Kim, Novack, Ross, and Teitelbaum, 2005; Wei, Kitaura, Zhou, Ross, and Teitelbaum, 2005).

It acts by stimulating the RANKL production and M-CSF (macrophage colony-stimulating

factor) from stromal cells and osteoblasts and amplify differentiation into osteoclasts

independently of RANK-RANKL signaling (Kawai, Stein, Perrien, and Griffin, 2012).

Moreover, the expression of RANK upregulates by synergistic effect of TNF-α and RANKL

(Komine, Kukita, Kukita, Ogata, Hotokebuchi, and Kohashi, 2001).

Page 30: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

11

1.6.2. Pro-inflammatory Cytokine: IL-1β

IL-1 is an inflammatory cytokine which is highly expressed in inflamed synovium in RA

(Symons, McDowell, Di Giovine, Wood, Capper, and Duff, 1988). Like TNF-α, it is produced

by several immune cells mainly by monocytes and macrophages (Chizzolini, Dayer, and

Miossec, 2009). IL-1 is more potent than TNF-α in causing cartilage destruction

(Vervoordeldonk and Tak, 2002). Previous studies have shown the important role of IL-1 on

bone metabolism by observing noticeable protection against structural damage offered by IL-

1 inhibition in an arthritis model (Joosten, Helsen, Saxne, van de Loo, Heinegård, and van den

Berg, 1999). Moreover, various animal models with IL-1 signaling deficiency have shown

increased bone density, cortical thickness and trabecular bone mass with reduced

osteoclastogenesis (Lee, Fujikado, Manaka, Yasuda, and Iwakura, 2010; Simsa-Maziel,

Zaretsky, Reich, Koren, Shahar, and Monsonego-Ornan, 2013).

1.7. RANK-RANKL-OPG in Bone Diseases

The RANK-RANKL-OPG axis regulates normal physiology of bone. Changes in interaction

and ratio of RANK-RANKL-OPG has significant role in bone diseases. Imbalance in the

coordination between activity and level of these molecules causes osteoclast formation and

activity to increase or decrease resulting in abnormal bone phenotypes such as osteoporotic or

osteopetrotic. Typical examples of such bone loss are associated with inflammatory diseases

such as periodontal disease, RA, and periprosthetic bone loss. Increased levels of RANKL as

compared to OPG are found to be observed in gingival tissue, prosthetic loosening, RA,

synovial-like soft tissue and soft tissues surrounding the damaging bone in cases of active

periodontal disease ( Haynes, Crotti, Potter, Loric, Atkins, Howie, and Findlay, 2001; Crotti,

Smith, Weedon, Ahern, Findlay, Kraan, Tak, and Haynes, 2002; Crotti, Smith, Hirsch,

Soukoulis, Weedon, Capone, Ahern, and Haynes, 2003; Holding, Findlay, Stamenkov, Neale,

Lucas, Dharmapatni, Callary, Shrestha, Atkins, and Howie, 2006; Haynes, Crotti, Weedon,

Slavotinek, Au, Coleman, Roberts‐ Thomson, Ahern, and Smith, 2008; Jiang, Li, Quan, Xiao,

Zhao, Wang, Liu, Gou, An, and Huang, 2015). Moreover, the increases RANKL expression

compared to OPG is associated with stimulated activity and differentiation of osteoclasts

(bone resorbing cells) (Haynes, Crotti, Potter, Loric, Atkins, Howie, and Findlay, 2001; Crotti,

Page 31: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

12

Smith, Hirsch, Soukoulis, Weedon, Capone, Ahern, and Haynes, 2003; Crotti, Smith, Findlay,

Zreiqat, Ahern, Weedon, Hatzinikolous, Capone, Holding, and Haynes, 2004; Holding,

Findlay, Stamenkov, Neale, Lucas, Dharmapatni, Callary, Shrestha, Atkins, and Howie, 2006;

Haynes, Crotti, Weedon, Slavotinek, Au, Coleman, Roberts‐ Thomson, Ahern, and Smith,

2008; Van Tuyl, Voskuyl, Boers, Geusens, Landewé, Dijkmans, and Lems, 2010) .

1.7.1. RANK/RANKL Pathway in Rheumatoid Arthritis

In the area of osteoclast research, identification of the receptor activator of nuclear factor- κB

(NF-κB) RANK and its ligand RANKL led to remarkable development (Kong, Feige, Sarosi,

Bolon, Tafuri, Morony, Capparelli, Li, Elliott, and McCabe, 1999). RANKL belongs to TNF

superfamily of cytokines and it was originally identified as a dendritic cell survival factor

produced by T cells activation (Hofbauer, Khosla, Dunstan, Lacey, Boyle, and Riggs, 2000).

In the presence of M-CSF, osteoclasts are produced after differentiation from hematopoietic

precursor cells in vitro after stimulation by RANKL (Fata, Kong, Li, Sasaki, Irie-Sasaki,

Moorehead, Elliott, Scully, Voura, and Lacey, 2000). RANKL leads to the activation of the

bone-resorbing activity of mature osteoclasts. RANKL binds to its transmembrane receptor

RANK that belongs to the TNF receptor superfamily. These receptors are expressed by mature

osteoclasts and dendritic cells as well as by osteoclast precursor cells of monocyte-

macrophage lineage (Neumann, Gay, and Müller‐ Ladner, 2005). These Osteoclast precursors

have been shown to exist in spleen in wild-type mice (Takahashi, Akatsu, Udagawa, Sasaki,

Yamaguchi, Moseley, Martin, and Suda, 1988). RANK stimulation results in interaction with

TRAF6 and GAB2 that leads to further activation of series of downstream intracellular

signals, finally stimulates activation of NF-κB and AP1 transcription factors (Grigoriadis,

Wang, Cecchini, Hofstetter, Felix, Fleisch, and Wagner, 1994; Gravallese, Manning, Tsay,

Naito, Pan, Amento, and Goldring, 2000; Lam, Takeshita, Barker, Kanagawa, Ross, and

Teitelbaum, 2000; Ruocco, Maeda, Park, Lawrence, Hsu, Cao, Schett, Wagner, and Karin,

2005; Wei, Kitaura, Zhou, Ross, and Teitelbaum, 2005) (Fig. 1.2). Osteoprotegerin (OPG) is

another important factor in this system. It is a soluble receptor of RANKL and related to TNF

receptor superfamily. Interaction of RANKL and OPG inhibits the RANKL binding to its

receptor RANK, thereby inhibiting RANKL activity. Kong et al reported that CD4+ T cells

activation can promote osteoclast differentiation through production of soluble or surface-

Page 32: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

13

bound RANKL. The study also demonstrated RANKL expression in the synovial tissue of

adjuvant-induced arthritic (AIA) rats by activated T cells. These studies revealed the central

role of RANKL produced by activated T lymphocytes and synovial fibroblasts in joint and

bone destruction in RA (Kong, Feige, Sarosi, Bolon, Tafuri, Morony, Capparelli, Li, Elliott,

and McCabe, 1999; Wong, Besser, Kim, Arron, Vologodskaia, Hanafusa, and Choi, 1999;

Hofbauer, Khosla, Dunstan, Lacey, Boyle, and Riggs, 2000).

1.7.2. RANK/RANKL Pathway and the Immune Response

RANKL has immunoregulatory functions and it is produced by spleen, thymus, activated T

cells, lymph nodes and lymphoid patches in the intestine. RANK is expressed in

hematopoietic precursors, mature T cells and surface of dendritic cells. As a result of RANKL

and RANK interaction, T lymphocytes after activation by dendritic cells undergo

proliferation. RANKL treatment stimulates the antigen specific primary T cells response and

it might be important for memory response of T lymphocytes. In addition to this, RANK-

RANKL interaction is also responsible for expression of CD-40 and IL-12 production in

dendritic cells (Anderson, Maraskovsky, Billingsley, Dougall, Tometsko, Roux, Teepe,

DuBose, Cosman, and Galibert, 1997; Wong, Rho, Arron, Robinson, Orlinick, Chao,

Kalachikov, Cayani, Bartlett, and Frankel, 1997; Kong, Feige, Sarosi, Bolon, Tafuri, Morony,

Capparelli, Li, Elliott, and McCabe, 1999; Josien, Li, Ingulli, Sarma, Wong, Vologodskaia,

Steinman, and Choi, 2000). In the light of these reports, RANKL/RANK system appears to

be important in the development of lymphoid tissue as well as key player in B and T

lymphocytes precursors’ maturation in bone marrow (Josien, Wong, Li, Steinman, and Choi,

1999; Kong, Yoshida, Sarosi, Tan, Timms, Capparelli, Morony, Oliveira-dos-Santos, Van,

and Itie, 1999). Study on human volunteers have reported the role of RANKL in the formation

of osteoclast like cells in the peripheral blood mononuclear cells (PBMCs) (Kotake, Udagawa,

Hakoda, Mogi, Yano, Tsuda, Takahashi, Furuya, Ishiyama, and Kim, 2001). Another study

showed that RANKL expression by activated T cells is able to control bone remodeling and

osteoclastogenesis both in vitro and in vivo (Josien, Li, Ingulli, Sarma, Wong, Vologodskaia,

Steinman, and Choi, 2000). Therefore, RANKL/RANK system might be responsible for bone

loss as a result of systemic activation of T cells. Under normal homeostasis, these T cells are

probably not required because studies on mice model lacking T cells showed normal

Page 33: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

14

development of bone (Nakashima and Penninger, 2003). Whereas, remodeling of bone

through RANKL system can be disturbed by chronic T cell activation (Vidal, Brandstrom,

Jonsson, and Ohlsson, 1998).

1.8. Overview of Treatment Strategies

Till late 1980s, the available options for the treatment of RA were limited. Most of these

agents were slow in action and some of them such as gold salts and penicillamine were

associated with serious toxicities. These agents were generally described as ‘go low, go slow’.

In the beginning, non-steroidal anti-inflammatory drugs (NSAIDs) were recommended for the

treatment continued for longer duration (1–2 years). If this treatment was not beneficial,

specific therapy with anti-rheumatic agents in smaller doses were initially recommended and

after a long trial period at each step advancement in the treatment regime was suggested. Anti-

rheumatic medications were never administered as combination therapies but only as single

therapies. This strategy of treatment is called pyramid approach in which the base consisted

of approaches that was applicable for all RA patients e.g. simple analgesics, physical therapy,

rehabilitation and above the basal treatment approach stands the NSAIDs; moving upward in

the pyramid is the treatment strategy that was appropriate only to a limited number of RA

patients including the use of DMARDs and finally at the top of the pyramid comes the

experimental therapies for RA such as plasmapheresis. Other than these treatment modalities,

Glucocorticoid (GC) injections and oral dose and rheumatologic surgeries are also included if

needed. This pyramid treatment strategy was revised in 1980s based on several key

observations. It was recognized that early stages of RA are characterized by irreversible

damage to the articular structures which can be decreased to some extent using DMARDs.

The main ideas in remodeling or inverting of pyramid treatment strategy were the employment

of DMARDs at earlier stages of RA, optimum dosing, escalating therapies and consideration

of combination of DMARDs (Wilske and Healey, 1989; Gremillion and Van Vollenhoven,

1998). In early 1990s, several studies reported the use of combination therapy. In this context,

Page 34: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

15

Figure 1.2. RANKL stimulates RANK-dependent signaling cascade. Interaction of RANK

and RANKL results in association of GAB2 and TRAF6 adaptor molecules. This complex

further leads to activation of p38 (MAPK)-MK2 pathway first, then it results in IKK2

phosphorylation causing dissociation of IκB from NF-κB. Then other family of transcription

factors c-fos and c-jun are activated, finally followed by c-src/PI3K/Akt signaling cascade.

CSF1R binds to its receptor MCSF and ERK activation are other key events in the

development of osteoclast. CD44 is a negative regulator that interfere directly with these

pathways (adapted from Schett, Hayer, Zwerina, Redlich, and Smolen, 2005).

Page 35: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

16

study reported that combination of hydroxychloroquine (HCQ), methotrexate (MTX) and

sulfasalazine (SSZ) were more effective than treatment with only methotrexate and the

combined effect of two other drugs (O'dell, Leff, Paulsen, Haire, Mallek, Eckhoff, Fernandez,

Blakely, Wees, and Stoner, 2002).

The main concern regarding the use of combined DMARDs was associated with toxicities

that need to be managed. In another study, the combination of cyclosporine A (CyA) and

MTX and was found to be superior to MTX alone tested in a randomized trial (Tugwell,

Pincus, Yocum, Stein, Gluck, Kraag, McKendry, Tesser, Baker, and Wells, 1995). However,

it is noteworthy that this strategy of combining drug effect was not found to be successful in

all the combination therapy trials. One such randomized trial reported that MTX + AZA was

less effective compared to MTX only and was also more toxic (Willkens, Urowitz, Stablein,

Mckendry, Berger, Box, Fiechtner, Fudman, Paul Hudson, and Marks, 1992). Some

uncontrolled observational experiments also demonstrated that combination therapies

including cytotoxic drugs were in some cases even more aggressive and are associated with

considerable toxicities (Wollenhaupt and Zeidler, 1997; McCarty and Carrera, 1982; Walters

and Cawley, 1988). Later on, liver toxicity was observed as a result of this practice (Weinblatt,

Dixon, and Falchuk, 2000). Therefore, close monitoring is required, if combination therapy is

used.

1.8.1. Non-Biological Therapies for Rheumatoid Arthritis

The non-biological treatment modalities mainly include NSAIDs, glucocorticoids (GCs), and

DMARDs.

1.8.1.1. Non-Steroidal Anti-Inflammatory Drugs (NSAIDS)

NSAIDs belong to a diverse group of medications that share a common mode of action but

are different structurally. They act by blocking the rate limiting enzyme cyclo-oxygenase

(COX) involved in the prostaglandins production. They are primarily used for the treatment

of pain, temporary aches and musculoskeletal conditions such as tendonitis and bursitis.

Sometimes these agents are used to treat gout and spondyloarthropathies as a long-term

medication. These are commonly used for the treatment of osteoarthritis but also used in the

RA treatment. As a main therapy, NSAIDs can only be used in mild cases of RA. However,

Page 36: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

17

these agents could be added in appropriate anti-rheumatic treatment to provide symptomatic

relief. The main side effects of NSAIDs are gastric problems that can aggravate to cause peptic

ulcers, gastritis, perforations and bleeding. Therefore, histamine 2-antagonists, proton-pump

antagonists, or misoprostol are usually given with NSAIDs. In order to avoid gastrointestinal

toxicity related to NSAIDs, COX-2 specific inhibitors were developed. Unfortunately, the use

of this class of drug was reduced due to unexpected high risk of cardiovascular pathologies.

1.8.1.2. Glucocorticoids (GCs)

The efficacy of GCs (corticosteroids) has been reported for suppression of inflammation in

many diseases including RA. In inflammatory condition, the long term use of glucocorticoids

at effective dosage is associated with many side-effects. Therefore, its use is restricted to

specific cases, for example, in life threatening conditions of RA. High-dose of GCs is used in

combination with strong immunosuppressive agents. In special circumstances, moderate

doses of GCs for short duration can be used as a bridging therapy while slow acting DMARDs

start their action. Low doses of GCs in combination with DMARDs therapy do not show an

effective anti-inflammatory response, however, two randomized clinical trials have reported

enhanced efficacy of DMARDs when given in combination with low-dose GCs and to some

extent showed mild protective effect in radiological damage (Svensson, Boonen, Albertsson,

Van der Heijde, Keller, and Hafström, 2005; Wassenberg, Rau, Steinfeld, and Zeidler, 2005).

In rheumatologic practice, intra-articular GC injections are commonly used and can be very

effective and safe if used appropriately. In Denmark, two clinical trials in RA patients were

conducted using multiple intra-articular GC injections that showed excellent recovery

(Hetland, Stengaard-Pedersen, Junker, Lottenburger, Hansen, Andersen, Tarp, Svendsen,

Pedersen, and Skjødt, 2008; Hørslev-Petersen, Hetland, Junker, Pødenphant, Ellingsen,

Ahlquist, Lindegaard, Linauskas, Schlemmer, and Dam, 2013).

1.8.1.3. Conventional DMARDs

Conventional DMARDs are diverse group of pharmacological agents with anti-rheumatic

properties. Its mode of action is still not completely clear. Some common features shared by

these include low cost, relieves symptoms, reasonable tolerability, but it takes long time to

show its effect usually weeks to months that’s why these are commonly referred as SAARDs

Page 37: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

18

(slow acting anti-rheumatic drugs) and these drugs are also associated with toxic side effects.

Therefore, careful monitoring is required during treatment.

In DMARDs, one of the most common and effective drug is methotrexate (MTX) and it is

used as a first-line therapy for RA. MTX was initially synthesized for the treatment of cancer.

It is an anti-metabolite and folate antagonist. In chronic conditions like psoriatic arthritis

(PsA), psoriasis and RA, it has shown efficacy and safety at low dosage. Later on, a study

suggested that the efficacy of MTX in RA is not due to its anti-metabolic property, but it

specifically enhances adenosine production which is an anti-inflammatory endogenous

mediator (Cronstein, Eberle, Gruber, and Levin, 1991; Cronstein, Naime, and Ostad, 1995,

1993). The side effects associated with MTX are mainly hepatic dysfunction, mouth ulcers,

myelosuppression, gastrointestinal symptoms and teratogenicity.

Sulphasalazine (SSZ) is another member of DMARD family of drugs, developed initially for

the RA treatment. It was assumed that RA developed because of some bacteria present in the

gut that are causing inflammatory changes. In 1940s, Karolinska Institute (Stockholm), Nanna

Svartz designed a molecule possessing anti-inflammatory and antibacterial properties. The

efficacy of SSZ was not recognized until post-world war chaos. It was rediscovered in the

1960s and found to be effective for RA and inflammatory bowel disease (IBD) treatment. SSZ

is recommended as a substitute to MTX in RA. In order to achieve greater efficacy, SSZ can

also be given in combination with MTX. SSZ is associated with several side effects such as

hepatic dysfunction, allergic reactions (sulfa allergy), myelosuppression and gastrointestinal

symptoms.

Hydroxychloroquine (HCQ) was originally discovered as an anti-malarial agent. However, it

was serendipitously found to possess anti-rheumatic properties as well. It is rarely used as

monotherapy because considered to be a weaker agent. HCQ in combination with SSZ and

MTX can be used as a triple therapy treatment strategy initiated by O’Dell et al (O'dell, Leff,

Paulsen, Haire, Mallek, Eckhoff, Fernandez, Blakely, Wees, and Stoner, 2002). HCQ is well-

tolerated with small chances of retinopathy.

Leflunomide is a well-known pyrimidine synthesis antagonist. Its risks, efficacy and side

effects are comparably similar to MTX (Weinblatt, Kremer, Coblyn, Maier, Helfgott, Morrell,

Page 38: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

19

Byrne, Kaymakcian, and Strand, 1999). It can be used as an alternative of MTX. Combined

treatment with leflunomide and MTX increases efficacy but can cause severe toxicity problem

(Kremer, Genovese, Cannon, Caldwell, Cush, Furst, Luggen, Keystone, Weisman, and

Bensen, 2002).

1.8.2. Biologics in the Treatment of RA

In 1975, the discovery of monoclonal antibodies by Kohler and Milstein (Köhler and Milstein,

1975) suggested a new field in the area of therapeutics. This development in the field of

medicine helped in the designing of new drugs with targeted biological effect for the relevant

pathology. Soon after its discovery, it entered the field of transplantation and oncology.

Biological therapies in RA first targeted T lymphocytes, but these treatments were found to

be either toxic (anti-CD52) or ineffective (anti-CD4) (Moreland, Pratt, Mayes, Postlethwaite,

Weisman, Schnitzer, Lightfoot, Calabrese, Zelinger, and Woody, 1995; Van Der Lubbe,

Dijkmans, Markusse, Nässander, and Breedveld, 1995; Isaacs, Manna, Rapson, Bulpitt,

Hazleman, Matteson, Clair, Schnitzer, and Johnston, 1996). In the meantime, TNF antagonists

were used for the first time in septic shock but it was also not successful ( Cohen and Carlet,

1996; Fisher Jr, Agosti, Opal, Lowry, Balk, Sadoff, Abraham, Schein, and Benjamin, 1996;

Reinhart, Wiegand-Lohnert, Grimminger, Kaul, Withington, Treacher, Eckart, Willatts,

Bouza, and Krausch, 1996; Clark, Plank, Connolly, Streat, Hill, Gupta, Monk, Shenkin, and

Hill, 1998). However, in vitro study done by Kennedy Institute in London suggested that TNF

antagonism would be effective for the RA patients treatment and they were successful in

convincing the company that developed first anti-TNF monoclonal antibodies for allowing

them to do trial (Brennan, Jackson, Chantry, Maini, and Feldmann, 1989). The trial ended up

with unexpected results and that led to the start of new era in the treatment strategy of RA

(Van Vollenhoven, 2015).

1.8.2.1. Overview of Current Biologic Therapies

Currently, nine biological agents have been approved for the RA treatment. Among these nine

agents, five are TNF antagonist and the rest have different mode of action. These biological

agents are summarized in table 1.1 (McInnes and Schett, 2011). Generally, they are called

DMARDs but they are differentiated from conventional DMARDs and referred as biological

Page 39: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

20

DMARDs. The biologics especially anti-TNF agents have fast action. Some RA patients on

anti-TNF therapy often reports improvement in their condition on the day of treatment or

afterwards. Such results were only seen previously with the use of very high dose of

glucocorticoids (GCs) or after two to three months of therapy with methotrexate (MTX). The

biologics are relatively safe and well tolerated but the long-term use of these blocking agents

were developing concerns regarding serious infections and other malignancies as a potential

consequence of their use for such a long period. It became clear that long term use of anti-

TNF therapy leads to the reactivation of latent tuberculosis in RA patients and increase in the

risk of skin cancer was also reported (Van Vollenhoven, 2015).

The use of these biologic DMARDs has brought revolution in the RA management, but there

are concerns related to its toxic effects. Inhibition of TNF blocks the signaling of over

expressed protein in RA but in this process it also inhibits signaling protein that is important

for the normal immune response that leads to increased risk of bacterial and fungal infections

and other opportunistic pathogens (Saag, Teng, Patkar, Anuntiyo, Finney, Curtis, Paulus,

Mudano, Pisu, and Elkins‐ Melton, 2008). Etanercept and infliximab were the first two anti-

TNF agents that were used in clinical practice (Van Vollenhoven, 2015). After these two

adalimumab, cetrolizumab and golimumab were used (Doran, Crowson, Pond, O'Fallon, and

Gabriel, 2002). Most of the clinical trials with the TNF-blockers have shown risk of serious

infections (Ruderman, 2012).

1.8.3. Non-Pharmacological Treatments for RA

Beside the use of pharmacological treatments, non-pharmacological mode of treatment

strategies play significant role in the management of RA. During the entire period of RA

treatment, physiotherapy is suggested for medical rehabilitation of RA patients. The goal of

non-pharmacological interventions is to alleviate the patient’s sufferings, to control pain and

maintain general well-being (Stenström and Minor, 2003). Adjustments in daily life at the

work place and in the home can provide benefits to the patients by occupational therapy.

Nutritional improvement can also serve benefits; however, the practice is rather limited.

Gluten-free and vegan diet is found to provide some improvement to the patients in one small

randomized study (Hafström, Ringertz, Spångberg, Von Zweigbergk, Brannemark, Nylander,

Page 40: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

21

Table 1.1. Approved biologic therapies of rheumatoid arthritis.

Name Class Target Structure

Adalimumab Cytokine inhibitor TNF-α Human monoclonal antibody

Certolizumab

pegol

Cytokine inhibitor TNF-α Pegylated humanized Fab

fragment of an anti–TNF-α

monoclonal antibody

Etanercept Cytokine inhibitor TNF-α TNF-α receptor–Fc fusion

Golimumab Cytokine inhibitor TNF-α Human monoclonal antibody

Infliximab Cytokine inhibitor TNF-α Chimeric monoclonal

antibody

Tocilizumab Cytokine inhibitor Interleukin-6

receptor

Humanized monoclonal

antibody

Anakinra Cytokine inhibitor Interleukin-1 Interleukin-1 receptor

antagonist

Rituximab Cell-depleting agent CD20 Chimeric monoclonal

antibody

Abatacept Cell-depleting agent CD80 and

CD86

CTLA4–Ig fusion protein

Page 41: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

22

Rönnelid, Laasonen, and Klareskog, 2001). In another study, the consumption of

‘Mediterranean diet’ showed considerable benefits to patients (Sköldstam, Hagfors, and

Johansson, 2003) and was generally acceptable for the long duration. Significant

cardiovascular health benefits were also observed. At some point during the disease period,

most patients require psychosocial support that can be provided by a psychologist or social

worker, support groups organized through patient associations’ clinics and hospitals. The

rheumatologist can also play the role of a psychologist to provide emotional support to the

patient (Van Vollenhoven, 2015).

1.9. Collagen Induced Arthritis (CIA) Rat Model

One of the most extensively studied animal models of RA is collagen induced arthritis (CIA).

The important feature of CIA model is that it mimics human arthritis with both immunological

and pathological features. CIA is basically an autoimmune disease affecting joints and require

immune function of both T and B cell to autologous type II collagen (CII) (Andersson and

Holmdahl, 1990). RA is not limited to the joint but also involves multiple tissues and complex

pathways. Therefore, it is important to choose an animal model that has similarity to the

human condition of RA so that it can be translatable to the human studies (McNamee,

Williams, and Seed, 2015). Collagen can be obtained from various sources including chick,

bovine, human and porcine. The overall response varies considerably with the strain and

conditions of injection (Andersson and Holmdahl, 1990). In a model of CIA, collagen is

responsible for the induction of immune response. It is useful for determining the efficacy of

drug with anti-arthritic potential in a controlled manner. This kind of model is also helpful for

exploring the role of specific genes involved in RA, through the use of knockout mice

(Vincent, Williams, Maciewicz, Silman, Garside, and Group, 2012). Strains of mice and rats

that are susceptible for CIA development are used for induction of immunity with type II

collagen emulsified either in Freund's complete adjuvant (CFA) or Freund's incomplete

adjuvant (IFA) (Trentham, 1982; Holmdahl, Andersson, Goldschmidt, Jansson, Karlsson,

Malmstrom, and Mo, 1989).

The histology of CIA bone and cartilage resembles that of RA in terms of bone and cartilage

erosion and infiltrating cells in synovial tissue (Nabozny, Bull, Hanson, Griffiths, Luthra, and

Page 42: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

23

David, 1994). Chronic pain is a noticeable feature associated with RA and providing pain

relief to RA patient is an important task. Animal models of arthritis are also used to assess

pain and for the investigation of action of analgesics. When evoked pain response is tested,

onset of CIA is characterized by increased sensitivity to mechanical and thermal stimuli,

making it a suitable model to test analgesics (Inglis, Notley, Essex, Wilson, Feldmann, Anand,

and Williams, 2007).

1.10. Compounds Used in the Current Study

In the present study, we have used N-(2-hydroxyphenyl) acetamide (NA-2), rutin and its gold

nanoparticles.

1.10.1. N-(2-Hydroxyphenyl) Acetamide (NA-2)

NA-2 is a derivative of acetaminophen (2-Acetamidophenol), which has been known as

antipyretic and analgesic agent. Acetaminophen has been used widely as an alternative to

aspirin because of less side effects but it has been reported to have nephrotoxic and

hepatotoxic effects due to conversion of drug by cytochrome P-450 dependent oxidase system

into reactive intermediate (Potter, Davis, Mitchell, Jollow, Gillette, and Brodie, 1973;

Mohandas, Duggin, Horvath, and Tiller, 1981; Patten, Thomas, Guy, Lee, Gonzalez,

Guengerich, and Yang, 1993). Classical NSAIDs have been used since long for the treatment

of RA but they only provide symptomatic relief and they also don’t protect the joint

destruction process (Santana-Sabagun and Weisman, 2001). Therefore, drugs with minimal

side effects over long term use and targeted mode of action are required. NA-2 is available

commercially and commonly known as 2-hydroxyacetamide, 2-acetamidophenol, O-

acetaminophenol, 2-acetylaminophenol and O-hydroxyacetanilide. After extensive literature

survey, we were not able to find much work on this compound, however in 2005, study by

Saeed and Saeed suggested the use of NA-2 as anti-platelet aggregating and anti-inflammatory

agent and they also reported that NA-2 is less toxic in comparison to aspirin or paracetamol

(Saeed and Saeed, 1975). NA-2 has been reported as potent anti-inflammatory and anti-

arthritic agent by our research group (Perveen, Hanif, Jawed, Jamall, and Simjee, 2014; Jawed,

Shah, Jamall, and Simjee, 2010; Jawed, Jamall, Shah, Perveen, Hanif, and Simjee, 2014) but

the possible mechanism behind its anti-arthritic potential is not clear yet. In the current study,

Page 43: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

24

we aim to investigate the mechanism of NA-2 anti-arthritic activity with specific emphasis

on its role in regulation of RANK-RANKL pathway.

1.10.2. Rutin

Rutin belongs to flavonoids group of compounds. Flavonoids are present in almost all foods

of plant origin (Hollman and Katan, 1997) and have shown numerous biological activities e.g.

anti-hypertensive, anti-oxidant, anti-platelet, anti-inflammatory, anti-neoplastic anti-

microbial, anti-viral etc. in both in vitro and in vivo models (Formica and Regelson, 1995). In

inflammatory animal models flavonoids have shown potent anti-inflammatory response (Di

Carlo, Mascolo, Izzo, and Capasso, 1999; Guardia, Rotelli, Juarez, and Pelzer, 2001).

Likewise, rutin has also shown potent therapeutic effects like anti-inflammatory, anti-oxidant,

anti-carcinogenic in various animal studies (La Casa, Villegas, De La Lastra, Motilva, and

Calero, 2000; Kamalakkannan and Prince, 2006; Bishnoi, Chopra, and Kulkarni, 2007; Khan,

Ahmad, Ishrat, Khuwaja, Srivastawa, Khan, Raza, Javed, Vaibhav, and Khan, 2009). In the

treatment of diseases like inflammatory bowel disease, osteoarthritis and rheumatoid arthritis,

the potential therapeutic role of rutin has been supported by various animal and human studies.

Study by Ostrakhovitch and Afanas’ev showed effective inhibitory effect of rutin on oxygen

radical overproduction in RA patients (Ostrakhovitch and Afanas’ev, 2001; Kauss, Moynet,

Rambert, Al-Kharrat, Brajot, Thiolat, Ennemany, Fawaz, and Mossalayi, 2008).

1.10.3. Rutin Gold Nanoparticles (Rutin-GNPs)

Nanoparticles are 1-100 nm size particles. There is recent interest in the use of these

nanoparticles in the treatment and diagnosis of different diseases, since the size of these

nanoparticles is similar to the biological molecules present in the human system like

nucleic acid and proteins and also similar to the virus structure (Wiesenthal, Hunter,

Wang, Wickliffe, and Wilkerson, 2011). In medicine, one of the major advantages of using

engineered nanoparticles is the targeted delivery to specific site of action. Drugs like

celecoxib, ibuprofen, and diclofenac have been investigated for osteoarthritis and have

shown significantly reduced level of inflammatory cytokines TNF-α and IL-6 in the RA

patients synovial fluid and higher doses of these drugs showed more promising anti-

inflammatory response in RA patients thus improving the quality of life (Gallelli, Galasso,

Page 44: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

25

Falcone, Southworth, Greco, Ventura, Romualdi, Corigliano, Terracciano, and Savino,

2013). Therefore, these drugs can be used in the form of nanoparticles. The nanodrugs

when given to the patients, release slowly towards their target site and low doses of

these nanodrugs are enough to produce its effect, therefore its use can prevent the

harmful side effects associated with high doses (Radad, Al-Shraim, Moldzio, and Rausch,

2012). These nanoparticles are also suitable for those drugs that are not soluble in water

and they can be designed to specifically inhibit or enhance immune response and even

avoid recognition by immune system (Cooper, 2010; Zolnik, Gonzalez-Fernandez,

Sadrieh, and Dobrovolskaia, 2010). Recent advances in technology has enabled

researchers to use these engineered nanoparticles for imaging, diagnosis of disease,

targeted delivery of drug and treatment at the same time (Sanvicens and Marco, 2008).

Limited research has been done in the area of targeted drug delivery in RA. Few examples

include the targeted MTX delivery using poly (lactic-co-glycolic acid) nanoparticles with CD-

64 antibody in vitro in RAW 264.7 cells (Das Neves, 2014). In comparison to free drug, the

MTX nanoparticles were found to be more effective (Zhou, Yan, Hu, Springer, Yang,

Wickline, Pan, Lanza, and Pham, 2014). In one study, IL-6 monoclonal antibody has been

investigated in CIA mice in which the antibody was targeted with tocilizumab loaded

hyaluronate gold nanoparticles (Lee, Lee, Bhang, Kim, Kim, Ju, Kim, and Hahn, 2014). It

was also reported that in CIA mice, small dose of MTX nanoparticles were far better in

therapeutic response than the high doses (Scheinman, Trivedi, Vermillion, and Kompella,

2011). Therefore, we aim to investigate the role of rutin-GNPs (rutin-GNPs) on RA

progression and the possible mechanism of action.

Page 45: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

26

1.11. Objectives

The current treatment strategies for RA involve lifelong treatment with serious side effects.

There is unmet need for the development of therapeutic agent that suppresses and cures the

disease process without exerting toxic effects. In the light of aforementioned role of pro-

inflammatory cytokines and RANKL/RANK pathway in arthritis, we planned to investigate:

1. The effect of NA-2, rutin, indomethacin + rutin and rutin-GNPs on the development of

arthritis in CIA rats.

2. The effect of these compounds on pro-inflammatory and oxidative stress markers i.e. IL-

1β, TNF-α, NO, PO and GSH in serum samples.

3. The role of these compounds in regulation of RANK/RANKL signaling cascade by

determining change in gene expression of RANKL, RANK and its downstream mediators

i.e. NF-κB, c-fos, c-jun and Akt by quantitative RT-PCR.

4. The change in protein expression of c-Fos, pAkt and iNOS in spleen tissue by

immunohistochemistry.

Page 46: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

27

CHAPTER 2

METHODOLOGY

Page 47: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

28

2.1. Animal Care

Female Wistar rats 160-200 gms were housed in the animal resource facility under pathogen

free conditions at controlled temperature (21ºC ± 2ºC) and humidity (55ºC ± 5ºC) and light-

dark cycle of 12:12 hour with free access of standard diet and drinking water. The ethical

guidelines set by the International Center for Chemical and Biological Sciences (ICCBS),

Scientific Advisory Committee on Animal Care, Use, and Standards, were followed.

2.2. Chemicals

Bovine type II Collagen (lyophilized) was purchased from Chondrex (Redmond, WA, USA).

Freund’s complete adjuvant, NA-2 and indomethacin were purchased from Sigma (St. Louis,

MO, USA). The Freund’s incomplete adjuvant was purchased from MP Biochemicals

(Aurora, OH, USA). Rutin and its gold nanoparticles were provided by our collaborators

(ICCBS, University of Karachi, Pakistan).

2.3. Collagen Suspension Preparation

Lyophilized bovine type II collagen (CII) was mixed with Freund’s complete adjuvant (CFA)

in 1:1 ratio to make final concentration of 2 mg/mL for initial dose and incomplete Freund’s

adjuvant (IFA) for booster dose.

2.4. Induction of Arthritis

A volume of 0.2 ml (0.2 mg) collagen CII suspension was injected subcutaneously (s.c.) at

the tail base. On the seventh day after injection of CFA-CII, 0.1 ml of suspension containing

CII-IFA was re-injected at the same site. Animals were kept under observation until symptoms

of arthritis were prominent. Compounds treatment was started on the day of arthritis induction.

2.5. Treatment Strategy

Animals were divided into groups as outlined in table 2.1. Indomethacin was used as the

reference drug. Doses of NA-2, Rutin and its gold nanoparticles were selected after

preliminary dose finding studies on acute carrageenan-induced inflammatory rat model.

Page 48: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

29

Table 2.1. Treatment regime used for the testing of compounds for anti-arthritic activity

in collagen induced arthritis model in rats.

Groups Doses Route of administration

Normal Control - -

Vehicle Control 0.5 mL/animal Intraperitoneal

Gold Nanoparticles only 0.5 mL/animal Intraperitoneal

NA-2 only 5 mg/kg Intraperitoneal

Rutin only 50 mg/kg Intraperitoneal

Arthritic Control - -

Arthritic Vehicle 0.5 mL/animal Intraperitoneal

Arthritic + Gold Nanoparticles 0.5 mL/animal Intraperitoneal

Indomethacin + Arthritis 5 mg/kg Intraperitoneal

NA-2 + Arthritis 5 mg/kg Intraperitoneal

Rutin-25 + Arthritis 25 mg/kg Intraperitoneal

Rutin-50 + Arthritis 50 mg/kg Intraperitoneal

Indomethacin + Rutin + Arthritis Indomethacin 2 mg/kg

and Rutin 25 mg/kg

Intraperitoneal

Rutin-GNPs +Arthritis 12.5mg/kg Intraperitoneal

Page 49: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

30

2.6. Clinical Assessment of Arthritis

During the course of in-vivo experiment, animals were observed for changes in body weight,

paw oedema and arthritic score.

2.6.1. Arthritic Score

Animals were kept under careful observation for monitoring the symptoms of arthritis. All the

immunized rats developed CIA, apparent by inflamed paw, after approximately 18-22 days.

The severity of arthritis in each paw was scored on a scale of 0-4 as shown in table 2.2

(Bakharevski, Stein-Oakley, Thomson and Ryan, 1998). The arthritic index was calculated as

a summated index derived for each rat yielding a maximum score of 16/ animal.

2.6.2. Hind Paw Volume Measurement

The severity of arthritis was analyzed by measurement of change in paw volume using

Plethysmometer (model 7140, Ugo Basile, Italy) on alternate days starting from day 0 till the

termination of the experiment. It considers three dimensional changes in the pattern of

swelling of individual limbs in comparison to diameter measurements of tibiotarsal joints.

2.6.3. Body Weight Measurement

Before the induction of arthritis (day 0) till the termination of the study (day 22), change in

body weight of all the animals was measured every alternate day by limiting its movement in

a restrainer of an appropriate size to determine the progression of disease. The average change

in body weight was determined by subtracting the individual weight recording on alternate

days from weight of day 0.

Page 50: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

31

Table 2.2. Scoring system used to assess severity of paw inflammation.

Score Condition

0 Normal, no signs of arthritis

1 swelling or redness of the paw or 1 digit

2 two joints involved

3 more than two joints involved

4 severe arthritis of the entire paw and all digits

Page 51: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

32

2.7. Histopathological Examination

On day 22, histological examination of specimens from the knee joints was done. The steps

involve in histology are mentioned below:

2.7.1. Fixation

The joint samples were immersed in 10% neutral buffered formalin overnight for fixation.

2.7.2. Decalcification

After fixation, the joints were immersed in decalcifying solution for almost four weeks in 10%

EDTA solution and then re-immersed in fixative to stop further decalcification.

2.7.3. Processing

Tissue processing involves multiple steps that are mentioned below:

2.7.3.1. Dehydration

Dehydration involves use of increasing strength of dehydrating agent (alcohol) that causes

slow removal of water thus causing minimal damage to the tissue. The joint samples were

immersed step by step in each solution for one hour at 42ºC starting from 70% alcohol, 90%

and finally the samples were kept in 100% alcohol for 60 min (three times). The samples were

then kept in 100% alcohol overnight at 42ºC.

2.7.3.2. Embedding

After dehydration the samples were immersed in melted paraffin at 66ºC in oven and paraffin

was changed three times and finally the samples were kept overnight in paraffin in the oven

at 66ºC. Next day the samples were taken out of the paraffin and transferred to the tissue

embedding mold and then melted paraffin was poured on the mold and the cassette was put

on top of the mold and allowed to cool down on cooling pad of the tissue embedding system

and finally after cooling and setting of the paraffin, the sample was taken out of the mold and

cassette was removed and kept at room temperature.

Page 52: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

33

2.7.3.3. Microtomy

The tissue sections were cut with 4 μm thickness using microtome. The sections were then

transferred into hot water bath (45ºC), and then taken on the slides and kept on hot plate at

45ºC overnight for drying.

2.7.3.4. Staining

The slides containing tissue sections were immersed in xylene for 1-2 minute and then

transferred to decreasing % of alcohol i.e., from 100%, 90% and 70% for 2 minutes each and

finally in water. The sections were then stained with hematoxylin for 1 minute then washed

with water and then stained with eosin for 1 minute and then again washed with water and

dehydrated with increasing concentration of alcohol (70%, 90%, 100%) and slides were

mounted and bone sections were then observed under microscope (CK X41, Olympus, Tokyo,

Japan). Images were captured and visualized using image analysis software (NIS element AR

3.0).

2.8. Inflammatory Markers Analysis

2.8.1. Reactive Oxygen Species (ROS) and Antioxidant Measurement

Reactive oxygen species, nitric oxide (NO), peroxide (PO) and antioxidant glutathione (GSH)

levels in serum samples of arthritic, normal and treatment groups were determined

colorimetrically by specific assay kits.

2.8.1.1 Nitric Oxide Determination (NO)

NO was measured using Quantichrome Nitric Oxide assay kit (BioAssay Systems, Hayward,

USA) by following the manufacturer’s protocol. The kit measures NO by reduction of nitrate

to nitrite. Briefly, the standard dilutions were prepared from 0-50 μM concentration. The

deproteination of serum samples was done by addition of 75 mM ZnSO4 and centrifuging at

14000 rpm for 5 mins. Supernatant was further treated with 55 mM NaOH and then glycine

buffer was added and incubation was done at room temperature for 15 mins with activated

cadmium granules (3 granules/sample) with intermittent shaking. Finally, test samples (100

μL) and standards were added into the 96-wells plate in triplicate. Reagent A and reagent B

Page 53: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

34

(50 μL/each) were added in each well and incubated for 5 mins at room temperature and then

absorbance was taken at 540 nm. The NO concentration was measured using standard curve.

Where n=dilution factor.

2.8.1.2. Peroxide Determination (PO)

Peroxide production was measured by QuantichromeTM peroxide assay kit (BioAssay systems,

Hayward, USA). Briefly, standard dilutions from 0-100 μM concentration were prepared.

Standards and blank (20 μL each) in duplicate and samples in quadruplicate were transferred

to 96-well plates. Working reagent A (WRa; 100 μL) was added to the standard wells, blank

and first two wells of each sample and in the last two wells, working solution B (WRb) was

added and then the plate was incubated for 30 mins at room temperature and OD was taken at

585 nm (ELISA reader, Spectra Max 340). The sample OD was compared with calibration

curve.

2.8.1.3. Glutathione Quantification (GSH)

Serum concentration of GSH in samples of arthritic, normal and test groups was measured by

5, 5’-dithiobis 2-nitrobenzoic acid (DNTB) method using QuantichromeTM Glutathione assay

kit (Bioassay Systems, Hayward, USA). Briefly, serum sample and reagent A (120 μL each)

were mixed together and centrifuged for 2 minutes at 14,000 rpm. Clear supernatant in a

volume of 200 µL and 100 µL of reagent B were added in duplicate into 96-wells plate. In the

blank wells 100 µL of distilled water and 100 µL of GSH standard (100 μM) were added into

the respective wells of standard. Final volume of 300 µL in each well was adjusted with

distilled water followed by incubation at room temperature for 25 mins. The OD was taken at

412 nm (ELISA reader, Spectra Max 340).

The concentration of GSH was calculated by standard curve as follows:

Concentration of NO (μM) = OD Sample – OD Blank x n

Slope

Conc of GSH (μM) = OD Sample – OD Blank 100 n

OD Standard – OD Blank

Page 54: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

35

2.9. Pro-Inflammatory Cytokines IL-1β and TNF-α Analysis

The Quantification of IL-1β and TNF-α was performed with commercially available ELISA

kits and all the steps were performed following manufacturer’s guidelines. The concentration

of cytokines was determined by plotting the standard curve on MicroCal origin program

(Micro Cal Inc. USA).

2.9.1. IL-1β Measurement

All the standard dilutions, samples and other reagents were prepared according to kit manual

(R&D systems, Minneapolis, USA). Briefly, the pre-coated microplate was used and assay

diluent (50 µL) was added to each well of microplate and then sample, control and standard

were added (50 µL each) into the plate. The plate was mixed gently followed by incubation at

room temperature for 2 hours. The samples were then aspirated and washed five times with

the wash buffer (400 µL/well) and then rat IL-1β conjugate (100 µL) was added to all the

wells and the plate was sealed and incubated for 2 hours at room temperature. 100 μL substrate

solution was added in all wells after plate washing followed by incubation for 30 mins in dark.

At the end, 100 µL stop solution was added. OD was taken at 450 nm.

2.9.2. TNF-α Measurement

The preparation of the reagents and samples was done following the manufacturer’s

instructions (Thermo Scientific, Waltham, USA). Briefly 50 µL of pretreatment buffer was

added to pre-coated 96 wells microplate. Serum samples and standards (50 µL each) were

added to each well in duplicate followed by incubation at room temperature for 1 hour. The

wells were washed with wash buffer three times with squirt bottle then biotinylated antibody

reagent was added to all the wells and plate was sealed and incubated for further 1 hour. The

plate was rewashed thrice and then Streptavidin-HRP reagent (100 µL) was added to each

well followed by incubation at room temperature for 30 minutes. Following incubation, the

plate was washed and then TMB substrate solution (100 µL) was added to all the wells and

incubated in dark at room temperature for 10 minutes. Finally, 100 µL stop solution was added

and OD was taken at 450nm.

Page 55: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

36

2.10. Real time Reverse Transcriptase Polymerase Chain

Reaction (Real Time RT-PCR)

At the end of each experiment, spleen tissues were extracted from all the rats and stored at -

80oC for gene expression studies.

2.10.1. RNA Isolation and Quantification

The total RNA isolation from spleen tissue was done by Trizol RNA extraction method, a

modified method originally developed in 1987 and proved to be reliable method for RNA

isolation form tissues (Chomczynski & Sacchi, 1987). The frozen tissues (10-20 mg) were

placed in clean DNase/RNase free tubes containing Trizol (1 mL) (Invitrogen, CA, USA) and

homogenized using Ultra-Turrax homogenizer (Ultra-Turrax T8; IKA, Germany). The tubes

were incubated at room temperature for 5 mins and chloroform (200 μL) was added to all the

tubes and after shaking incubated for 3 mins at room temperature and then tubes were

centrifuged at 4ºC for 15 minutes at 12000 ×g. After centrifugation, total RNA present in the

upper aqueous phase was carefully shifted to a new tube and 500 μL isopropanol was added

into the tube. The tubes were incubated for 10 minutes followed by centrifugation at 4oC at

12,000 ×g for 10 mins. The supernatant was discarded and RNA pellets were mixed with 1

ml ethanol (75%) and centrifuged at 7500 ×g for 5 mins at 4oC. Supernatant was discarded

and pellet was air dried for 10 mins. Finally, the pellet was re-suspended in RNase free water

and quantification and purity of RNA was analyzed using nanodrop 2000 UV-Visible

spectrophotometer (Thermo Scientific Inc.) and then stored at -70ºC for further use.

2.10.2 The cDNA Synthesis

The cDNA was synthesized by reverse transcription of total RNA using Revert Aid TM First

strand cDNA Synthesis Kit (Thermo Scientific, Waltham, USA). The cDNA synthesis was

done using manufacturer’s protocol. Briefly, 1μg of RNA was used to make cDNA. The RNA

and primer mixture was made by adding the components in 0.5 mL tube in amounts shown in

the table 2.3. The mixture was incubated for 5 mins at 65ºC. The cDNA synthesis mix (Table

2.4) was added to the RNA/Primer mixture. The mixture (random hexamer primer) was then

Page 56: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

37

incubated for 5 mins at 25ºC and then for 60 mins at 42ºC. The reaction was stopped by

incubation at 70ºC for 5 mins and the cDNA was stored at -70ºC.

2.10.3. Real time Reverse Transcriptase-PCR Amplification of cDNA

The cDNA was then amplified according to manufacturer’s protocol using Maxima SYBR

green master mix (2X) (Table 2.5). Briefly, 2 μL of diluted cDNA (1:10) was used as a

template for real time PCR. Negative control without reverse transcriptase enzyme was used

to confirm the absence of genomic DNA contamination. GAPDH (glyceraldehyde-3-

phosphate dehydrogenase) gene was used as a reference housekeeping gene. The final reaction

mixture was placed in preheated real time PCR and the reaction details are mentioned in table

2.6. The list of primer sequences for the corresponding genes, primer concentration and

annealing temperature is mentioned in table 2.7.

2.11. Immunohistochemistry for c-Fos, iNOS and pAkt in Spleen

Tissue

The antibodies used for immunohistochemical analysis of c-Fos, iNOS and pAkt, in Spleen

tissues are given in table 2.8. After the development of full blown arthritis, the animals were

sacrificed. The spleen tissues were perfused with chilled PBS and fixed with 10% formalin

for 24 hrs. The samples were then dehydrated with increasing concentration of alcohol and

embedded in paraffin and tissue sectioning was done using microtome and the sections were

transferred to properly labeled poly-L-lysine coated slides. For immunostaining, the labeled

slides were rehydrated by transferring the slides step by step in xylene 100% (20-30 mins) →

100% alcohol (10 mins) → 90% alcohol (5 mins) → 70% alcohol (10 mins) → H2O (10 mins).

After that PBS was poured on top of sections and the slides were incubated at 42ºC in oven

for 15 mins in humid chamber. After incubation, the tissues were soaked with Roti-

immunoblock working solution (Carl Roth, Karlsruhe, Germany) and the slides were

incubated in oven at 42ºC for 30 mins. After blocking the tissue sections were incubated with

primary antibodies (1:100 dilution) of respective markers using blocking solution and

incubated at 42ºC for 1 hour. After incubation, the slides were washed with PBS (three times)

followed by incubation with secondary antibody (1:200 dilutions) at 42ºC for 45 mins-1 hour.

Page 57: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

38

Table 2.3. Composition of RNA/Primer mixture.

Component Volume

Total RNA (1 μg) 2 μL

Random Hexamer Primer 1 μL

Nuclease Free Water 9 μL

Total 12 μL

Table 2.4. Composition of cDNA synthesis mixture.

Component Volume

5X Reaction Buffer 4 μL

Ribolock RNase Inhibitor (20U/μL) 1 μL

10mM dNTP Mix 2 μL

Revert Aid M-MuLV RT (200U/μL) 1 μL

Page 58: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

39

Table 2.5. Real time RT- PCR reaction mixture recipe.

Component Volume (μL)

Maxima SYBR Green (2X) 12.5 μL

Forward primer (10μM) 0.5 μL (0.5 μM)

Reverse Primer (10μM) 0.5 μL (0.5 μM)

Template DNA 2 μL

Nuclease free water to 25 μL

Total 25 μL

Table 2.6. Steps involved in real time RT-PCR reaction.

Step Temperature (ºC) Time No. of Cycles

Initial denaturation 95 2 mins 1

Denaturation 95 15 secs

Annealing 60 30 secs 40

Extension 72 30 secs

Page 59: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

40

Table 2.7. Primer sequences, annealing temperature and expected product size.

Genes Accession.

No

Primer Sequence

(Forward/Reverse Primer)

Annealing

Temperature

(Tm)

Product

Size (bp)

RANK NM_001

271235.1

5’-A A C T C C A A C T C A A C G G A T G G-3’ 59.97 ºC 124

5’-T G G G A A G G C C T A T G C T G T A G-3’ 60.23 ºC

RANKL NM_057

149.1

5’-A C A G C G C T T C T C A G G A G T T C-3’ 59.75 ºC 136

5’-G A C T T T A T G G G A A C C C G A T G-3’ 59.24 ºC

c-Fos NM_022197.2

5’-C C T G C C T C T T C T C A A T G A C C-3’ 59.80 ºC 111

5’-G A T G C C G G A A A C A A G A A G T C-3’ 59.68 ºC

c-Jun NM_021

835.3

5’-C C G G A G A A G A A G C T C A C A A G-3’ 60.13 ºC 111

5’-C T C T G G G T C A G G A A A G T T G C-3’ 59.84 ºC

NF-κB

p65

NM_199

267.2

5’-G A A C T T G T G G G G A A G G A C T G-3’ 58.10 ºC 128

5’-C T A A T G G C T T G C T C C A G G T C-3’ 58.33 ºC

Akt1 NM_033

230.2

5’-A C T C A T T C C A G A C C C A C G A C-3’ 59.97 ºC 169

5’-A C C A C G T T C T T C T C G G A G T G-3’ 60.30 ºC

GAPDH NM_017

008.4

5’-A T G A C T C T A C C C A C G G C A A G-3’ 60.13 ºC 136

5’-T A C T C A G C A C C A G C A T C A C C-3’ 59.86 ºC

Page 60: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

41

The slides were washed and stained with DAPI (1:10000 dilutions in PBS) and incubated for

10 mins at room temperature for nuclear staining. Finally, the slides were washed and mounted

with mounting media and protected from light. Negative controls were also processed by

simply elimination of the primary antibody from the procedure in order to determine the

specificity of primary antibody. Upright multichannel fluorescent microscope (Nikon Eclipse

90i, Nikon Corporation, Japan) was used for the visualization of sections and pictures were

taken using camera (Nikon, DXM1200C) and the images were analyzed using image analysis

software (NIS Element AR 3.0).

2.11.1. Image Analysis

The analysis of images was done using ImageJ software (National Institutes of Health, USA).

The Data was obtained from three sections per rat and presented as means ± S.E.M.

2.12. Statistical Analysis

The SPSS version 22 was used for the analysis of data. All the data was analysed using one-

way ANOVA and Bonferroni’s Post Hoc test was used to determine the mean differences

between different groups. The values equal or less than the level of P < 0.05 were considered

statistically significant.

Page 61: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

42

Table 2.8. List of antibodies for immunohistochemistry.

Markers Primary Ab Company Secondary Ab Company

c-Fos c-Fos Rabbit

Polyclonal IgG

(sc-52)

Santa Cruz

Biotechnology

Cy3 goat antibody

IgG (H+L)

(A10520)

Invitrogen

pAkt Affinity Purified

Rabbit IgG

(AF887)

Invitrogen Cy3 goat antibody

IgG (H+L)

(A10520)

Invitrogen

iNOS iNOS Rabbit

polyclonal

(Ab15323)

Abcam Cy3 goat antibody

IgG (H+L)

(A10520)

Invitrogen

Page 62: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

43

CHAPTER 3

RESULTS

Page 63: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

44

3.1. Clinical Parameters of Collagen Induced Arthritis

Animals in different treatment groups were carefully monitored on alternate days for severity

of arthritis using standard arthritic scoring criteria (Table 2.2), change in body weight and paw

volume. The effects of test compounds on clinical signs of arthritis are discussed in the

subsequent sections.

3.1.1. Effect of NA-2 on Clinical Parameters

3.1.1.1. Arthritic Score

The severity of arthritis was measured by analysing the mean arthritis severity score. Which

was determined by adding the score of individual paws of each rat. The animals in all the

groups showed no change in the mean arthritic score till day 10 (Fig. 3.1). However, from day

10 onward, inflammation was observed in arthritic control and NA-2 treated animals either in

one or both hind paws. In contrast, the reference drug indomethacin did not show any sign of

inflammation till day 12. On day 14, a significantly increased mean arthritic score (p < 0.0001)

was found in the untreated arthritic group compared to the non-arthritic animals and this

increase was persistent till the end of the study (day 22).

Next, we compared the NA-2 treated animals with the untreated arthritic group and observed

that it was able to reduce the arthritic score from day 14. This reduction in arthritic score of

the treated group was observed till the end of the study (p < 0.0001). The post hoc test revealed

a significant reduction in the mean arthritic score of indomethacin treated animals (p < 0.05),

on day 14, in comparison to NA-2 treatment, however this difference was not observed from

day 16 onwards.

Since the compound NA-2 was soluble in 5% DMSO, therefore we also set a vehicle control

group for arthritic and non-arthritic animals. The statistical analysis of the data gathered from

this control group did not reveal any significant difference from normal control or arthritic

control groups.

Page 64: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

45

3.1.1.2. Body Weight

The mean change in the body weight of the animals in various groups is shown in figure 3.2.

A gradual increase in the body weights of the animals in all the groups was observed until day

8. However, on day 10 onwards, the arthritic control animals demonstrated decrease in their

mean change in body weight and from day 12 onwards, this decrease was significant (p <

0.05) compared to the non-arthritic group. Both NA-2 (5 mg/kg) and indomethacin (5 mg/kg)

treatment groups showed increase in body weight from day18 onwards but compared to

untreated arthritic group, only indomethacin exhibited significant increase (p < 0.05) in mean

change in body weight on day 22 but NA-2 group remain statistically non-significant. The

Bonferroni’s post hoc test showed non-significant difference within the non-arthritic control

groups, arthritic control groups and between NA-2 and indomethacin treatment groups.

3.1.1.3. Paw Oedema

The change in the volume of both right and left paw was determined on day 0 till day 22 (Fig.

3.3). The non-arthritic control group exhibited only slight change in paw volume till the end

of the study because of increase in body weight but this increment was non-significant. In

comparison to the normal control animals, the untreated arthritic animals revealed significant

increase in both right and left paw volume (p < 0.0001 and p < 0.05 for right and left paw)

from day 12 and remain significant till the end of the study (p < 0.0001). From day 12

onwards, in comparison to the arthritic control group, indomethacin group started to show

significant reduction in both right paw (p < 0.0001) and left paw volume (p < 0.05) and this

difference remain significant till the end of the study (p < 0.0001). Similarly, from day 12,

NA-2 treatment group showed significant decrease in the right paw volume (p < 0.001) and

from day 14 in the left paw volume (p < 0.05). This mean change in paw volume of both right

and left paw remains significant in comparison to the arthritic control rats till day 22 (p <

0.0001). The statistical analysis demonstrated non-significant difference between the

treatment groups.

Page 65: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

46

Figure 3.1. Effect of NA-2 on mean arthritic severity score in rats induced with

collagen induced arthritis. All values are shown as mean ± S.E.M. (12 animals/group).

The untreated arthritic animals demonstrated significant increase in the mean arthritic

score from day 14 onwards (#p < 0.0001) as compared to the normal control rats.

However, in comparison to the arthritic control rats, NA-2 and indomethacin treated

animals exhibited significant decrease in the mean score from day 14 (*p < 0.0001).

Figure 3.2. Effect of NA-2 on mean change in body weight. The values are mean ±

S.E.M. post induction of arthritis (12 animals/group). The arthritic control rats exhibited

significant reduction in body weight compared to the normal control animals from day

12 onwards (#p < 0.05). Whereas, on day 22, the control drug indomethacin treated

animals demonstrated significant increase in body weight compared to the arthritic

control animals (*p < 0.05).

#

#

* *

**

0

2

4

6

8

10

12

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Mea

n

Art

hri

tic

Sco

re

Days after Immunization

Normal Control Vehicle Control

NA-2 Only Arthritic Control

Arthritic + Vehicle Arthritic + Indomethacin (5 mg/kg)

Arthritic + NA-2 (5 mg/kg)

#

#

*

-20

-15

-10

-5

0

5

10

15

20

25

30

35

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Mea

n C

han

ge

in B

od

y W

eig

ht

(gm

s)

Days after Immunization

Normal Control Vehicle Control

NA-2 Only Arthritic Control

Arthritic + Vehicle Arthritic + Indomethacin (5 mg/kg)

Arthritic + NA-2 (5 mg/kg)

Page 66: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

47

Figure 3.3. Effect of NA-2 on mean change in hind paw volume after arthritis

induction. Each value represents mean ± S.E.M. (12 animals/group). Compared to the

normal control animals, the untreated arthritic control animals exhibited significant

increase in both right (#p < 0.0001) and left paw volume (#p < 0.05) from day 12 onwards

and remain significant till day 22 (#p < 0.0001 for right paw and ## p < 0.0001 for left paw).

From day 12 onwards, compared to the arthritic control, indomethacin (5mg/kg) treated

group showed significant decrease in both right paw volume (***p < 0.0001) and left paw

volume (*p < 0.05) and remain significantly less till day 22 (***p < 0.0001). In contrast

NA-2 group showed significant reduction in right paw volume on day 12 (**p < 0.001,)

and from day 14 left paw volume (*p < 0.05) and the significance persist till day 22 for

both right and left paw compared to the arthritic control group (***p < 0.0001).

#

#

*** ***** ***

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Ch

an

ge

in

Rig

ht

Paw

Volu

me

(mL

)

Days after Immunization

Normal Control Vehicle Control

NA-2 Only Arthritic Control

Arthritic + Vehicle Arthritic + Indomethacin (5 mg/kg)

Arthritic + NA-2 (5 mg/kg)

#

##

* ***

* ***

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Ch

an

ge

in L

eft

Paw

Volu

me

(mL

)

Days after Immunization

Normal Control Vehicle Control

NA-2 Only Arthritic Control

Arthritic + Vehicle Arthritic + Indomethacin (5 mg/kg)

Arthritic + NA-2 (5 mg/kg)

B

A

Page 67: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

48

3.1.2. Effect of Rutin on Clinical Parameters

3.1.2.1. Arthritic Score

Figure 3.4 demonstrates the mean arthritic severity score of non-arthritic and arthritic animals.

All the animals look normal with no prominent change in mean arthritic severity score till day

10. On day 12, compared to the normal control rats, the untreated arthritic animals exhibited

increase in score that was significantly higher on day 14 onwards (p < 0.0001). From day 14

onwards, in comparison to the untreated arthritic control animals, the score of indomethacin

(5 mg/kg) treatment group exhibited marked reduction (p < 0.0001). Treatment of arthritic

rats with rutin (25 mg/kg) showed gradual increase in the arthritic score from day 12 till day

20, and on day 22 the score decreased slightly in comparison to the untreated arthritic animals.

A gradual increase in arthritic score of the rutin (25 mg/kg) treated arthritic group was lower

than the arthritic control animals and on day 14, it was significantly less than the arthritic

control rats (p < 0.01). On day 14 onwards, arthritic score of Indomethacin + rutin (2 mg/kg

+ 25 mg/kg) treated groups demonstrated a significantly reduced mean arthritic score (p <

0.0001). The arthritic animals treated with rutin (50 mg/kg) also demonstrated a significant

decrease in the arthritic score from day 16 (p < 0.05) and the score continue to decrease

significantly till day 22 (p < 0.001). Next the statistical analysis was done to determine the

differences within the treatment groups and it showed that in comparison to rutin (25 mg/kg),

from day 14 onwards, indomethacin treatment group (p < 0.0001) and day 18 onwards

indomethacin + rutin treatment group (p < 0.05) showed significant decrease in the mean

arthritic severity score. Rutin (50 mg/kg) also demonstrated reduced arthritic score but it was

statistically non-significant in comparison to rutin (25 mg/kg) group. The difference within

the non-arthritic control animals was non-significant.

3.1.2.2. Body Weight

The animals showed increment in the average body weight till day 8 (Fig. 3.5). When

compared to the non-arthritic control rats, the untreated arthritic animals exhibited significant

reduction (p < 0.05) in mean change in their body weights from day 12 onwards and it

continued to decrease significantly till day 22 (p < 0.0001). The indomethacin treated group

demonstrated significant increase (p < 0.05) in average change in body weight in comparison

Page 68: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

49

Figure 3.4. Effect of rutin on mean arthritic score. The values represent mean ± S.E.M.

of 12 animals/group. Day 14 onwards, the arthritic control rats showed significantly

increased arthritic score compared to normal control animals (#p < 0.0001). The

indomethacin treatment reduced the paw oedema (τp < 0.0001) from day 14 onward. On

day 14, the rutin (25 mg/kg) exhibited significantly less (**p < 0.01) arthritic score as

compared to the arthritic control rats, day 16 onwards non-significant difference was

observed. On day 14 onwards, indomethacin + rutin (2 mg/kg + 25 mg/kg) showed

significant decrease in score (**p < 0.01 and τp < 0.0001). From day 16 onwards, rutin (50

mg/kg) score continue to decrease significantly till the end of the study (***p < 0.001).

Figure 3.5. Mean change in body weight of arthritic and non-arthritic animals. The

values are mean ± S.E.M. (12 animals/group). Day 12 onwards, the untreated arthritic

animals showed significantly reduced body weight (#p < 0.05) compared to the normal

animals and continue to decrease significantly till day 22 (##p < 0.0001). The indomethacin

treated group showed significant increase in body weight compared to the arthritic control

group on day 22 (*p < 0.05). Rutin (50 mg/kg) and indomethacin + rutin (2 mg/kg + 25

mg/kg) exhibited non-significant increase in average body weight. Whereas, rutin (25

mg/kg) showed decrease in body weight compared to arthritic control animals.

#

#

ττ

**

*

*****

τ0

5

10

15

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Mea

n A

rth

riti

c S

core

Days After Immunization

Normal Control Rutin Only

Arthritic Control Arthritic + Indomethacin (5 mg/kg)

Arthritic + Rutin (25 mg/kg) Arthritic + Rutin (50 mg/kg)

Arthritic + Indomethacin + Rutin (2 mg/kg + 25 mg/kg)

# ##

*

-20

0

20

40

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Mea

n c

han

ge

in b

od

y w

eig

ht

(gm

s)

Days after immunization

Normal Control Rutin Only

Arthritic Control Arthritic + Indomethacin (5 mg/kg)

Arthritic + Rutin (25 mg/kg) Arthritic + Rutin (50 mg/kg)

Arthritic + Indomethacin + Rutin (2 mg/kg + 25 mg/kg)

Page 69: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

50

to the arthritic control rats on day 22. Day 16 onwards, rutin (50 mg/kg) and day 18 onwards

indomethacin + rutin (2 mg/kg + 25 mg/kg) treated animals showed gradual but non-

significant increase in body weight till the end of the study. Rutin (25 mg/kg) group showed

gradual decrease in body weight from day 10 till day 22. Bonferroni’s post hoc test determined

the differences between the treatments groups and it showed that in comparison to the

indomethacin group, the rutin (25 mg/kg) group showed significant decrease (p < 0.01) in

mean change in body weight. There was no significant difference within the normal and

arthritic control animals.

3.1.2.3. Paw Oedema

The mean change in paw volume of both right and left paw is shown in figure 3.6. From day

12, clear inflammation was observed in the arthritic control rats which was significantly higher

in comparison to the normal control group in both right paw (p < 0.0001) and left paw (p <

0.05). It was observed that the indomethacin (5 mg/kg) and indomethacin + rutin (2 mg/kg +

25 mg/kg) treated groups exhibited reduced paw volume from day 12 onwards. When

compared with the arthritic control animals, this reduction was significantly different both in

right paw volume (p < 0.0001) and left paw volume (p < 0.05). The arthritic animals treated

with 50 mg/kg of rutin demonstrated a gradual increase in their paw volume from day 12 till

day 16, however this increase was markedly less than arthritic control group. From day 16

paw volumes begin to reduce gradually and on day 22 the reduction was significant in

comparison to the untreated arthritic animals (p < 0.001). The comparison of treatment groups

showed that from day 12 till the end of the study, the indomethacin (5 mg/kg) and

indomethacin + rutin group (2 mg/kg + 25 mg/kg) showed significant reduction in their right

(p < 0.0001) and left paw volumes (p < 0.01) compared to rutin (25 mg/kg). On day 12, rutin

(50 mg/kg) in comparison to the rutin (25 mg/kg), exhibited marked decrease in the right paw

volume (p < 0.0001) and remain significant till day 22 (p < 0.01) treatment group.

Page 70: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

51

Figure 3.6. Mean change in paw volume following treatment with rutin. All values are

mentioned as mean ± S.E.M. of n=12/group. A significant increase in both right and left

hind paw volume of arthritic control rats was detected from day 12 onwards (##p < 0.0001

and #p < 0.05) compared to normal control group. Day 12 till the end of the study,

indomethacin and indomethacin + rutin treatment group showed significant decrease in

both right (***p < 0.0001) and left paw volume (*p < 0.05). Rutin (50 mg/kg) also showed

significant decrease in right paw volume on day 12 and 22 (***p < 0.0001 and **p < 0.001)

and left paw volume from day 20 onwards (*p < 0.05 and **p < 0.001).

##

##

*** ******

**

***

***

0

0.5

1

1.5

2

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Ch

an

ge

in R

igh

t P

aw

Volu

me

(mL

)

Days after Immunization

Normal Control Rutin Only

Arthritic Control Arthritic + Indomethacin (5 mg/kg)

Arthritic + Rutin (25 mg/kg) Arthritic + Rutin (50 mg/kg)

Arthritic + Indomethacin + Rutin (2 mg/kg + 25 mg/kg)

#

##

* ***

***

* ***

0

0.5

1

1.5

2

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Ch

an

ge

in L

eft

Paw

Volu

me

(mL

)

Days after Immunization

Normal Control Rutin Only

Arthritic Control Arthritic + Indomethacin (5 mg/kg)

Arthritic + Rutin (25 mg/kg) Arthritic + Rutin (50 mg/kg)

Arthritic + Indomethacin + Rutin (2 mg/kg + 25 mg/kg)

B

A

Page 71: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

52

3.1.3. Effect of Rutin-GNPs on Clinical Parameters of RA

3.1.3.1. Arthritic Score

For initial 10 days, all the animals showed score of 0. However, on day 12, except

indomethacin (5 mg/kg) group, the arthritic control group and rutin-GNPs treated groups (12.5

mg/kg) showed slight increase in their mean arthritic score which was significantly high (p <

0.0001, Fig. 3.7) from day 14 onwards compared to the non-arthritic control rats. The score

of the arthritic control group gradually increase with time and remain significantly higher (p

< 0.0001) till the end of the study. In comparison to the arthritic control group, both

indomethacin (p < 0.0001) and rutin-GNPs (p < 0.001) treatment exhibited significantly

reduced mean arthritic score on day 14 and remain significant till the end of the study (p <

0.0001 and p < 0.01 for indomethacin and rutin-GNPs respectively). The indomethacin score

in comparison to the rutin-GNPs treatment group, was significantly less from day 14 till day

16 (p < 0.0001; p < 0.05) and then the difference became non-significant till the end of the

study. There was also no significant difference between controls included in this study.

3.1.3.2. Body Weight

Figure 3.8 depicts the mean change in body weight with S.E.M. The mean change in body

weight of all the animals showed increasing trend till day 8. Compared to normal control, the

untreated arthritic animals, started to show significant reduction (p < 0.05) in body weight

from day 12 onwards and this pattern was consistent till day 22 (p < 0.0001). Indomethacin

demonstrated significant increase in body weight on day 22 (p < 0.05) but rutin-GNPs

treatment group showed no significant increase compared to the arthritic control animals. The

statistical analysis to find the difference between treatment groups showed no change and non-

significant difference was found within the normal and arthritic control animals.

3.1.3.3. Paw Oedema

The mean change in hind paws volume of all the groups is shown in figure 3.9. In comparison

to the normal control group, the arthritic control group showed significant increase in the right

and left paw volume from day 12 onwards (p < 0.0001 and p < 0.05 for right and left paw

respectively). In contrast, from day 12 onwards, compared to the arthritic control group,

Page 72: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

53

indomethacin (5 mg/kg) treatment group, showed significant reduction in mean change in

volume of both hind paws (p < 0.0001 and p < 0.05 for right and left paw respectively) and

this pattern of significance continued till the end of the experiment (p < 0.0001). Whereas,

rutin-GNPs (12.5 mg/kg) treatment group showed significant decrease in right paw volume

on day 12 and 22 (p < 0.0001 and p < 0.05 respectively) and from day 14 onwards, in the left

paw volume (p < 0.05 and p < 0.0001). The post hoc analysis also determined the difference

between the treatment groups and it showed that the indomethacin (5 mg/kg) treatment group

demonstrated significant reduction in mean change in right paw volume from day 20 onwards

(p < 0.05). Whereas, the left paw volume showed no significant difference compared to rutin-

GNPs (12.5 mg/kg) treatment group. The difference within non-arthritic control groups was

statistically non-significant.

Figure 3.7. Effect of rutin-GNPs on arthritic score. All the values are shown as mean ±

S.E.M. (12 animals/group). In comparison to the normal control animals, on day14

onwards, the untreated arthritic rats exhibited significant increase in mean arthritic severity

score (#p < 0.0001). Whereas, indomethacin and rutin-GNPs treatment group showed

significant reduction in mean arthritic score from day 14 (***p < 0.0001 and **p < 0.001)

and remain significant till day 22 (***p < 0.0001 and *p < 0.01).

#

#

******

***

0

2

4

6

8

10

12

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Mea

n A

rth

riti

c S

core

Days after Immunization

Normal Control Gold Nanoparticles Only

Arthritic Control Arthritic + Gold Nanoparticles

Arthritic + Indomethacin (5 mg/kg) Arthritic + Rutin-GNPs (12.5 mg/kg)

Page 73: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

54

Figure 3.8. Effect of rutin-GNPs on mean change in body weight. The values are

mentioned as mean ± S.E.M. (12 animals/group). In comparison to the normal control rats,

the untreated arthritic control animals exhibited significant reduction in body weight from

day 12 onwards (#p < 0.05) and increase in significance till day 22 (##p < 0.0001). The

indomethacin treatment group showed significant increase in weight on day 22 compared

to the arthritic control group (*p < 0.05).

# ##

*

-20

-15

-10

-5

0

5

10

15

20

25

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Mea

n C

han

ge

in B

od

y W

eig

ht

(gm

s)

Days after Immunization

Normal Control Gold Nanoparticles Only

Arthritic Control Arthritic + Gold Nanoparticles

Arthritic + Indomethacin (5 mg/kg) Arthritic + Rutin-GNPs (12.5 mg/kg)

Page 74: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

55

Figure 3.9. The mean change in paw volumes following treatment with rutin-GNPs.

The values are mentioned as mean ± S.E.M. The animals in the arthritic control

demonstrated significant increase in mean change in both right and left paw volume from

day 12 onwards compared to the normal control group (#p < 0.05 and ##p < 0.0001). In

contrast, indomethacin (5 mg/kg) treatment showed significant reduction in paw volume

from day 12 onwards compared to the arthritic control group (**p < 0.0001 and *p < 0.05

for right and left paw). Whereas, rutin-GNPs group showed significant reduction in right

paw volume on day 12 and 22 (**p < 0.0001 and *p < 0.05) and from day 14 onwards in

the left paw (*p < 0.05 and **p < 0.0001) respectively.

## ##

****

**

*

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Ch

an

ge

in R

igh

t P

aw

Volu

me

(mL

)

Days after Immunization

Normal Control Gold Nanoparticles Only

Arthritic Control Arthritic + Gold Nanoparticles

Arthritic + Indomethacin (5 mg/kg) Arthritic + Rutin-GNPs (12.5 mg/kg)

# ##

* ***

**

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

0 2 4 6 8 1 0 1 2 1 4 1 6 1 8 2 0 2 2

Ch

an

ge

in L

eft

Paw

Volu

me

(mL

)

Days after Immnunization

Normal Control Gold Nanoparticles Only

Arthritic Control Arthritic + Gold Nanoparticles

Arthritic + Indomethacin (5 mg/kg) Arthritic + Rutin-GNPs (12.5 mg/kg)

B

A

Page 75: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

56

3.2. Histological Analysis of Knee Joints

When the experiment was terminated (day 22), knee joint samples were collected from all the

included groups and H & E staining was performed for analysis of changes in joint

architecture. The histological characteristics associated with the pathogenesis of RA included

granulation tissue proliferation, synovial infiltration with lymphocytes and other

inflammatory cells and collapse of articular surface and pannus formation. The histological

scores were also determined for arthritic and non-arthritic animals indicating 4 as the

maximum score for the highest degree of inflammation and joint erosion (Table 3.1). The

representative joint histology and their respective scores of all the arthritic and non-arthritic

animals treated with the studied compounds is mentioned in the subsequent section.

3.2.1. NA-2 Treatment

An intact articular cartilage with negligible synovial infiltration can be seen in H&E stained

knee joint sections of the normal control animals (Fig. 3.10). In contrast, arthritic control

group showed prominent synovial infiltration and damaged articular cartilage. The

indomethacin and NA-2 treatments were able to reduce the infiltration in the synovium as well

as reduced the further destruction of articular cartilage in comparison to that observed in

arthritic group receiving no other treatment.

3.2.2. Rutin Treatment

In contrast to the arthritic control group which showed destruction of articular cartilage and

highly infiltrated synovial membrane, the treatment groups, i.e. the rutin (50 mg/kg),

indomethacin and indomethacin + rutin (2 mg/kg + 25 mg/kg) demonstrated less synovial

infiltration and almost negligible tissue damage (Fig. 3.11). In contrast, the treatment of

arthritic animals with rutin (25 mg/kg) was unable to reverse the inflammation and almost

same tissue damage was seen which was prominent in untreated arthritic animals.

3.2.3. Rutin-GNPs Treatment

The knee joints histopathological analysis of rutin-GNPs (12.5 mg/kg) treated group showed

less infiltration of synovium and intact cartilage (Fig. 3.12).

Page 76: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

57

Table 3.1. Histological scoring system adopted to monitor the histological changes

in arthritic and non-arthritic rats.

Group Knee Joint

Inflammation Erosion

Normal Control 0 0

Arthritic Control 4 4

Arthritis + Indomethacin 2 1

Arthritis + NA-2 2 1

Arthritis + Rutin (25 mg/kg) 4 3

Arthritis + Rutin (50 mg/kg) 2 1

Arthritis + Indomethacin + rutin (2

mg/kg + 25 mg/kg)

2 1

Arthritis + Rutin-GNPs 2 1

The score was determined on 0-4 scale. Where 0 score indicates absence of inflammation

or erosion in the joints. Whereas, score of 2-3 indicated mild to moderate tissue erosion

and inflammation, score 4 demonstrate severe inflammation, extensive articular destruction

and severe bone erosion.

A

Page 77: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

58

Figure 3.10. Effect of NA-2 treatment on joint destruction in CIA rats. (A) Normal

control; lack of lymphocyte proliferation and intact articular cartilage can be observed. (B)

Arthritic control; pronounced proliferation of lymphocytes (arrow 2) and erosion of articular

cartilage (arrow 1) as compared to the normal control can be seen. (C and D) Indomethacin

and NA-2 treated group; prominent decrease in inflammatory changes can be observed as

compared to the arthritic control. Where AC: articular cartilage; SM: synovial membrane; P: periarticular tissue.

A B

C D

1 2

P P

AC AC

AC

SM

AC

Page 78: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

59

Figure 3.11. Effect of rutin treatment on Knee joints sections from CIA rats. (A)

Normal control; negligible synovial infiltration with lymphocytes and intact articular

cartilage can be observed. (B) Arthritic control; prominent lymphocytic proliferation

(arrow 2) and collapse of articular surface (arrow 1) as compared to the normal control can

be seen. (C) Indomethacin treated group; reduced inflammatory response can be observed

compared to arthritic control. (D) rutin (25 mg/kg) treatment group showed almost similar

inflammatory changes compared to the arthritic control rats. (E and F) The rutin (50 mg/kg)

and Indomethacin + rutin treatment group showed normal joint architecture with no visible

signs of cartilage erosion and less synovial infiltration can be seen compared to the

untreated arthritic control group. Where AC: articular cartilage; SM: synovial membrane;

P: periarticular tissue.

A B

C D

E F

2 1 SM

AC

AC

AC

P P

AC

2

Page 79: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

60

Figure 3.12. Effect of rutin-GNPs treatment on Knee joints sections from CIA rats .

(A) Normal control; lack of lymphocyte proliferation and damage to articular cartilage can

be clearly observed. (B) Arthritic control; pronounced lymphocytic proliferation (arrow 2)

and collapse of articular structure (arrow 1) can be seen compared to the normal control.

(C and D) Indomethacin and rutin-GNPs treated group showed no signs of bone damage

and less synovial infiltration can be seen as compared to the arthritic control group. Where AC: articular cartilage; SM: synovial membrane; P: periarticular tissue.

A B

C D

2

SM 1

P P

AC

AC

AC

Page 80: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

61

3.3. Measurement of Oxidative Stress Markers in Serum

The oxidative stress parameters include nitric oxide (NO), peroxide (PO) that are pro-

inflammatory markers and glutathione (GSH), which is an endogenous antioxidant system

were determined from serum samples of all the animals at the end of in vivo study. The effect

of the compounds used in the study on these parameters is mentioned below.

3.3.1. Effect of NA-2 Treatment

3.3.1.1. Serum Nitric Oxide (NO)

The level of serum NO in the untreated arthritic control rats was significantly increased (p <

0.0001) as compared to the normal control rats (Fig. 3.13). The NA-2 and indomethacin

treatment significantly reduced serum NO concentration compared to the arthritic control

group (p < 0.0001). The statistical analysis revealed non-significant difference between the

treatment groups and also within the arthritic and non-arthritic control groups studied.

3.3.1.2. Serum Peroxide (PO)

The concentration of peroxide in serum sample of arthritic and non-arthritic animals is

mentioned in figure 3.13. The arthritic control rats exhibited significant increase (p < 0.0001)

in the PO concentration as compared to the normal control animals. The PO level was

significantly reduced in the treatment groups, when compared to the arthritic control animals

(p < 0.0001). However, difference between the treatment groups and similarly within the non-

arthritic and arthritic control animals was non-significant.

3.3.1.3. Glutathione (GSH)

The figure 3.13 shows the concentration of serum GSH. Statistical analysis demonstrated a

significantly decreased level of serum GSH in the untreated arthritic control animals as

compared to the normal control animals (p < 0.0001). A significant increase in the GSH level

was noted in the arthritic groups treated with NA-2 and indomethacin (p < 0.0001) as

compared to the arthritic control group.

Page 81: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

62

Figure 3.13. Effect of NA-2 on serum NO, PO and GSH level in arthritic and non-

arthritic animals. Data is shown as mean ± S.E.M. for 12 animals/group. Animals in

the arthritic group demonstrated significantly higher level of NO, PO and significantly

low level of GSH concentration compared to the normal control animals (#p < 0.0001).

Indomethacin and NA-2 treatment groups showed significantly reduced level of NO and

PO and significantly high GSH level (*p < 0.0001) as compared to the untreated arthritic

control animals.

#

* *

0

2

4

6

8

10

12

14

16

18

20

Normal Control Vehicle Control NA-2 Only Arthritic

Control

Arthritic +

Vehicle

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + NA-

2 (5 mg/kg)

NO

Con

cen

trati

on

g/m

L)

Treatment Groups

#

* *

0

5

10

15

20

25

30

Normal ControlVehicle Control NA-2 Only Arthritic

Control

Arthritic +

Vehicle

Arthritic +

Indomethacin

(5mg/kg)

Arthritic +NA-

2 (5 mg/kg)

PO

Con

cen

trati

on

(n

g/m

L)

Treatment GroupsB

#

*

*

0

1

2

3

4

5

6

7

8

Normal Control Vehicle Control NA-2 Only Arthritic

Control

Arthritic +

Vehicle

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + NA-

2 (5 mg/kg)

GS

H C

on

cen

tra

tio

n (

mg

/dL

)

Treatment GroupsC

A

Page 82: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

63

3.3.2. Effect of Rutin on Oxidative Stress Parameters

3.3.2.1. Serum Nitric Oxide (NO)

Compared to the normal control group, significantly increased serum NO level was detected

in the arthritic control rats (p < 0.0001; Fig. 3.14). Whereas, the serum level of NO in

indomethacin (5 mg/kg, p < 0.0001), rutin (50 mg/kg, p < 0.0001), and indomethacin + rutin

(2 mg/kg + 25 mg/kg, p < 0.001) treated groups was significantly reduced in comparison to

arthritic control animals. It was also noted that treatment of arthritic animals with rutin (25

mg/kg) had no effect on the measured NO concentration and it was almost same as that of the

arthritic control animals.

3.3.2.2. Serum Peroxide (PO)

The arthritic control group showed significantly high PO level in serum compared to the

normal control group (p < 0.0001; Fig. 3.14). The indomethacin (5 mg/kg), rutin (50 mg/kg)

and indomethacin + rutin (2 mg/kg + 25 mg/kg) showed significantly reduced PO level in

comparison to the arthritic control group (p < 0.0001). Whereas, rutin (25 mg/kg)

demonstrated non-significant difference in PO concentration as compared to the arthritic

control rats. The statistical analysis of treatment groups showed significant reduction in PO

concentration (p < 0.0001) in comparison to the rutin (25 mg/kg). The difference within the

normal and arthritic control animals was statistically non-significant.

3.3.2.3. Serum Glutathione (GSH)

A prominent decrease in the serum glutathione concentration was detected in the arthritic

control rats compared to the normal control animals (p < 0.0001; Fig. 3.14). All the treatment

groups except rutin (25 mg/kg), showed significantly high concentration of GSH in

comparison to the untreated arthritic animals (p < 0.0001 for indomethacin (5 mg/kg) and

rutin (50 mg/kg) and p < 0.001 for indomethacin + rutin (2 mg/kg + 25 mg/kg) treatment

group respectively). When treatment groups were compared, the indomethacin (5 mg/kg) and

rutin (50 mg/kg) treatment groups showed significantly high GSH level (p < 0.0001 and p <

0.05) in comparison to the rutin (25 mg/kg). The difference within the arthritic and normal

control groups was statistically non-significant.

Page 83: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

64

3.3.3. Effect of Rutin-GNPs on Oxidative Stress Parameters

3.3.3.1. Serum Nitric Oxide (NO)

Figure 3.15, shows concentration of NO in the non-arthritic and arthritic rats. In comparison

to the normal control rats, the untreated arthritic control animals revealed significantly high

NO concentration (p < 0.0001). In contrast, indomethacin and rutin-GNPs showed significant

decrease in NO concentration in comparison to the arthritic control group (p < 0.0001). Non-

significant difference was found between the treatment groups.

3.3.3.2. Serum Peroxide (PO)

In the arthritic control rats, the concentration of PO was significantly high compared to the

non-arthritic control rat (p < 0.0001; Fig. 3.15). Whereas, in comparison to the arthritic control

group, the indomethacin and rutin-GNPs treatment group demonstrated significant reduction

in the PO level (p < 0.0001). The difference between the treatment groups was non-

significant. Similarly, no difference was found within the normal and arthritic control group.

3.3.3.3. Effect of Rutin-GNPs on Serum Glutathione (GSH)

Figure 3.15 demonstrates the level of GSH in the serum samples of untreated arthritic and

non-arthritic groups. The GSH concentration of arthritic control animals was found to be

significantly decreased as compared to the normal control rats (p < 0.0001). The indomethacin

(5 mg/kg) and rutin-GNPs (12.5 mg/kg) showed significantly high level of GSH as compared

to the arthritic control animals (p < 0.0001). The statistical analysis exhibited non-significant

difference between the treatment groups and within the arthritic and non-arthritic control

group.

Page 84: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

65

Figure 3.14. The NO, PO and GSH concentration measured following rutin

treatment. The values are shown as mean ± S.E.M. for n=12/group. Significant

increase in NO, PO concentration and significant reduction in GSH level was found in

arthritic control animals compared to the normal control (#p < 0.0001). In contrast, the

indomethacin (5 mg/kg) and rutin (50 mg/kg) showed significant reduction in NO and

PO concentration and significant increase in GSH concentration (**p < 0.0001).

However, indomethacin + rutin (2 mg/kg + 25 mg/kg) treatment group also showed

significant reduction in NO and PO level and significant increase in GSH level

compared to the arthritic control animals (*P < 0.001 for NO and GSH and **p <

0.0001 for PO).

#

**

** *

0

2

4

6

8

10

12

14

16

18

20

Normal

Control

Rutin Only Arthritic

Control

Arthritic +

Indomethacin

(5 mg/kg)

Arthritic +

Rutin (25

mg/kg)

Arthritic +

Rutin (50

mg/kg)

Arthritic +

Indomethacin +

Rutin (2 mg/kg

+ 25 mg/kg)

NO

Con

cen

trati

on

g/m

L)

Treatment Groups

#

** ** **

0

5

10

15

20

25

30

Normal Control Rutin Only Arthritic

Control

Arthritic +

Indomethacin

(5 mg/kg)

Arthritic +

Rutin (25

mg/kg)

Arthritic +

Rutin (50

mg/kg)

Arthritic +

Indomethacin +

Rutin ( (2

mg/kg + 25

mg/kg)

PO

Con

cen

trati

on

(n

g/m

L)

Treatment GroupsB

#

****

*

0

1

2

3

4

5

6

7

8

Normal Control Rutin Only Arthritic

Control

Arthritic +

Indomethacin (5

mg/kg)

Arthritic +

Rutin (25

mg/kg)

Arthritic +

Rutin (50

mg/kg)

Arthritic +

Indomethacin +

Rutin (2 mg/kg

+ 25 mg/kg)

GS

H C

on

cen

trati

on

(m

g/d

L)

Treatment GroupsC

A

Page 85: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

66

Figure 3.15. Serum NO, PO and GSH concentration measured after treatment of

rutin-GNPs. The values are mentioned as mean ± S.E.M. for 12 animals/group. A

significant increase in NO and PO level and significantly reduced GSH level was detected

in the untreated arthritic control rats compared to the normal control rats (#p < 0.0001).

However, the indomethacin and rutin-GNPs treatment showed significant reduction in the

NO and PO concentration and significant increase in GSH concentration in comparison

to the arthritic control animals (*p < 0.0001).

#

**

0

5

10

15

20

25

Normal Control Gold Nanoparticles

Only

Arthritic Control Arthritic +

Goldnanoparticles

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin-

GNPs (12.5 mg/kg)

NO

Con

cen

trati

on

g/m

L)

Treatment GroupsA

#

* *

0

5

10

15

20

25

30

Normal Control Gold Nanoparticles

Only

Arthritic Control Arthritic + Gold

nanoparticles

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin-

GNPs (12.5 mg/kg)

PO

Con

cen

trati

on

(n

g/m

L)

Treatment GroupsB

#

**

0

1

2

3

4

5

6

7

8

Normal Control Gold Nanoparticles

Only

Arthritic Control Arthritic + Gold

Nanoparticles

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin-

GNPs (12.5 mg/kg)

GS

H C

on

cen

trati

on

(m

g/d

L)

Treatment GroupsC

Page 86: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

67

3.4. Measurement of Pro-Inflammatory Cytokines in Serum

TNF-α and IL-1β concentration was determined in both arthritic and non-arthritic animals.

The subsequent sections demonstrate the concentration of both these cytokines following

treatment with test compounds.

3.4.1. Effect of NA-2 on Serum IL-1β and TNF-α Concentration

The concentration of both IL-1β and TNF-α in serum samples of arthritic and non-arthritic

rats are mentioned in figure 3.16. In comparison to the non-arthritic control animals, IL-1 β (p

< 0.0001) and TNF- α (p < 0.01) level was significantly increased in the untreated arthritic

control animals. However, indomethacin treatment (5 mg/kg) exhibited significant decrease

in IL-1β (p < 0.0001) and TNF-α (p < 0.001) concentration compared to the arthritic control

group. The NA-2 (5 mg/kg) treatment group also showed significant decrease in both the

cytokine level (p < 0.0001 for IL-1β and p < 0.01 for TNF- α) respectively. There was no

significant difference within the arthritic, non-arthritic and treatment groups.

3.4.2. Effect of Rutin on IL-1β and TNF-α Concentration

The arthritic control group demonstrated significantly high serum concentration of IL-1β (p

< 0.0001) and TNF-α (p < 0.01) compared to the normal control rats (Fig. 3.17). However,

indomethacin (5 mg/kg), rutin 50 (mg/kg) and indomethacin + rutin (2 mg/kg + 25 mg/kg)

demonstrated significantly reduced IL-1β concentration (p < 0.0001) and TNF-α

concentration (p < 0.001 for indomethacin and p < 0.01 for rutin-50 mg/kg and indomethacin

+ rutin treatment groups, respectively) in comparison to the arthritic control rats. IL-1β and

TNF-α concentration of rutin (25 mg/kg) treatment group was non-significant compared to

the arthritic control animals. The comparison of treatment groups showed that indomethacin

(5 mg/kg, p < 0.0001); rutin (50 mg/kg, p < 0.001) and indomethacin + rutin (2 mg/kg + 25

mg/kg, p < 0.0001) showed significantly reduced level of IL-1β compared to the rutin (25

mg/kg). Whereas, TNF- α concentration of only indomethacin (5 mg/kg) treatment group was

significantly reduced (p < 0.012). The difference within the normal control groups was non-

significant.

Page 87: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

68

Figure 3.16. Effect of NA-2 on IL-β and TNF-α concentration. Each bar represents

mean ± S.E.M. (12 animals/group) A) A significantly high level of IL-1β was observed

in the arthritic control animals (#p < 0.0001) compared to the normal control animals.

Whereas, NA-2 and indomethacin treatment showed significantly reduced level as

compared to the arthritic control group (*p < 0.0001) B) The arthritic control rats

showed significantly high level of TNF-α in the serum samples compared to the normal

control rats (#p < 0.01). Whereas, the serum level of TNF-α in the indomethacin (**p <

0.001) and NA-2 (*p < 0.01) treated animals was significantly low in comparison to the

arthritic control animals.

#

**

0

20

40

60

80

100

120

Normal Control Vehicle Control NA-2 Only Arthritic

Control

Arthritic +

Vehicle

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + NA-

2 (5 mg/kg)

IL-1

βC

on

cen

trati

on

(p

g/m

L)

Treatment Groups

#

** *

0

50

100

150

200

250

300

350

400

450

500

Normal Control Vehicle Control NA-2 Only Arthritic

Control

Arthritic +

Vehicle

Arthritic +

Indomethacin (5

mg/kg)

Arthritic +NA-

2 (5 mg/kg)

TN

F-α

Con

cen

trati

on

(p

g/m

L)

Treatment GroupsB

A

Page 88: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

69

Figure 3.17. Effect of rutin on IL1-β and TNF-α concentration. A) The serum

concentration of IL-1β of all the rats is shown as mean ± S.E.M. (12 animals/group).

Compared to the normal control rats, the untreated arthritic control rats exhibited marked

increase in the level of IL-1β (#p < 0.0001), whereas the treatment groups except rutin (25

mg/kg) showed significantly decreased level of IL- β (*p < 0.0001) B) The arthritic control

group showed significant increase in the TNF-α concentration in comparison to the normal

control group (#p < 0.01). Compared to the arthritic control rats, except rutin (25 mg/kg)

that showed non-significant reduction in TNF-α level, all the treatment groups i.e.

indomethacin, rutin (50 mg/kg) and indomethacin + rutin (2 mg/kg + 25 mg/kg) showed

significantly decreased TNF-α level (**p < 0.001 and *p < 0.01).

#

*

* *

0

20

40

60

80

100

120

Normal Control Rutin Only Arthritic Control Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin

(25 mg/kg)

Arthritic + Rutin

(50 mg/kg)

Arthritic +

Indomethacin +

Rutin (2 mg/kg +

25 mg/kg)

IL-1

βC

on

cen

trati

on

(p

g/m

L)

Treatment GroupsA

#

*** *

0

50

100

150

200

250

300

350

400

450

500

Normal Control Rutin Only Arthritic Control Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin

(25 mg/kg)

Arthritic + Rutin

(50 mg/kg)

Arthritic +

Indomethacin +

Rutin (2 mg/kg

+ 25 mg/kg)

TN

F-α

Con

cen

trati

on

(p

g/m

l)

Treatment GroupsB

Page 89: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

70

3.4.3. Effect of Rutin-GNPs on IL-1β and TNF-α Concentration

A significantly high level of IL-1β (p < 0.0001) and TNF-α (p < 0.01) in the untreated arthritic

rats was found as compared to the normal control rats (Fig. 3.18). In the treatment groups, i.e.

indomethacin (5 mg/kg) and rutin-GNPs (12.5 mg/kg), significant reduction in both IL1-β (p

< 0.0001) and TNF-α concentration (p < 0.001 and p < 0.01 for indomethacin and rutin-GNPs

treatment group, respectively) was found in comparison to the arthritic control rats. The

statistical analysis showed no difference within the normal control, arthritic control and the

treatment groups.

3.5. Gene Expression Studies Using Quantitative Real time RT-

PCR

The quantitative gene expression analysis of RANKL pathway i.e. RANK, RANKL, NF-κB,

c-fos, c-jun and Akt was done in the spleen tissue samples of the arthritic and non-arthritic

rats. The primer sequences of the genes studied is mentioned in table 2.7.

3.5.1. Effect of NA-2 Treatment

The change in the expression of RANK, RANKL, NF-κB, c-fos, c-jun and Akt genes

following treatment with NA-2 is mentioned in Figure 3.19. The arthritic control group

showed significantly high gene expression of all the aforementioned genes (p < 0.0001) in

comparison to the normal control animals. In comparison to the arthritic control rats, a

significant decrease in the fold change of RANK, RANKL, c-fos and c-jun gene expression

was detected in the indomethacin and NA-2 treatment groups (p < 0.0001). Similarly, a

significantly reduced expression of NF-κB was observed in the indomethacin (p < 0.001) and

NA-2 treatment groups (p < 0.0001). Significant reduction in the Akt gene expression was

also observed in the NA-2 (p < 0.01) and indomethacin (p < 0.0001) treatment groups. The

statistical analysis of all the genes mentioned above showed non-significant difference

between the treatment groups and similarly within the arthritic and non-arthritic control

groups.

Page 90: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

71

Figure 3.18. Effect of rutin-GNPs on IL-1β and TNF-α concentration. The values are

shown as mean ± S.E.M. (12 animals/group). A) The serum IL-1β concentration of arthritic

control animals showed marked increase compared to the normal control animals (#p <

0.0001) and the indomethacin and rutin-GNPs treatment groups showed significantly

reduced IL-1β concentration (*p < 0.0001). B) The arthritic control rats showed

significantly high serum TNF-α concentration as compared to the normal control animals

(#p < 0.01). The indomethacin (5 mg/kg) and rutin-GNPs (12.5 mg/kg) treatment group

demonstrated significantly reduced level of TNF-α (**p < 0.001 and *p < 0.01)

respectively.

#

* *

0

20

40

60

80

100

120

Normal Control Gold Nanoparticles

Only

Arthritic Control Arthritic + Gold

Nanoparticles

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin-

GNPs (12.5

mg/kg)

IL-1

βC

on

cen

trati

on

(p

g/m

l)

Treatment GroupsA

#

***

0

50

100

150

200

250

300

350

400

450

500

Normal Control Gold Nanoparticles

Only

Arthritic Control Arthritic + Gold

Nanoparticles

Arthritic +

Indomethacin (5

mg/kg)

Arthritic + Rutin-

GNPs (12.5 mg/kg)

TN

F-α

Co

nce

ntr

ati

on

(p

g/m

l)

Treatment GroupsB

Page 91: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

72

3.5.2. Effect of Rutin Treatment

The effect of rutin on genes of RANKL pathway i.e. RANK, RANKL, NF-κB, c-fos, c-jun

and Akt genes is shown in figure 3.20. The arthritic control group as compared with the normal

control group showed significant increase in the expression of all the genes mentioned above

(p < 0.0001). Whereas the indomethacin (5 mg/kg), rutin (50 mg/kg) and indomethacin + rutin

(2 mg/kg + 25 mg/kg) treatment groups showed significant decrease in RANK gene

expression as compared to the untreated arthritic control animals (p < 0.0001). Rutin (25

mg/kg) showed non-significant change in comparison to the arthritic control group in all the

genes studied. The comparison of treatment groups showed a marked suppression in gene

expression in indomethacin, rutin (50 mg/kg) and indomethacin + rutin (2 mg/kg + 25 mg/kg)

treatment groups (p < 0.0001) compared to rutin (25 mg/kg). The difference within the normal

and arthritic control groups was statistically non-significant.

The expression of RANKL in indomethacin, rutin (50 mg/kg), and indomethacin + rutin (2

mg/kg + 25 mg/kg) treatment groups (p < 0.0001) was significantly down regulated as

compared to the arthritic control animals. Whereas, the comparison of treatment groups

revealed that in rutin (50 mg/kg) and indomethacin (5 mg/kg) group significant reduction in

the RANKL gene expression (p < 0.0001) was observed in comparison to rutin (25 mg/kg)

treatment group. Whereas, indomethacin + rutin (2 mg/kg + 25 mg/kg) also demonstrated

significant reduction in gene expression (p < 0.001) compared to rutin (25 mg/kg).

The NF-κB gene expression in the treatment groups except rutin (25 mg/kg) showed

significant reduction in comparison to the arthritic control group (p < 0.001 for indomethacin

and p < 0.0001 for rutin-50 mg/kg and indomethacin + rutin (2 mg/kg + 25 mg/kg) groups

respectively). The difference within the treatment groups was statistically non-significant.

The c-fos gene expression in all the treatment groups i.e. indomethacin, rutin (50 mg/kg) and

indomethacin + rutin (2 mg/kg + 25 mg/kg), showed significantly reduced expression in

comparison to the arthritic control group (p < 0.0001). Compared to the rutin (25 mg/kg)

treatment group, significant reduction in the gene expression was observed (p < 0.001 for

indomethacin and p < 0.0001 for rutin-50 mg/kg and indomethacin + rutin (2 mg/kg + 25

mg/kg treatment groups).

Page 92: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

73

The c-jun gene expression in the indomethacin (5 mg/kg), rutin (50 mg/kg) and indomethacin

+ rutin (2 mg/kg + 25 mg/kg) treatment groups showed significant reduction (p < 0.0001) in

comparison to the arthritic control animals. Rutin (25 mg/kg) showed non-significant

difference in comparison to the arthritic control group. In comparison to rutin (25 mg/kg)

significant decrease in the indomethacin (p < 0.0001) and rutin (50 mg/kg p < 0.0001) and

indomethacin + rutin (2 mg/kg + 25 mg/kg, p < 0.01) treatment group was observed.

In the case of Akt gene expression, the indomethacin (p < 0.0001), rutin (50 mg/kg, p < 0.01)

and indomethacin + rutin (2 mg/kg + 25 mg/kg, p < 0.01) treated groups showed significantly

decreased expression as compared to the arthritic control animals. The rutin (50 mg/kg, p <

0.05) and indomethacin (5 mg/kg, p < 0.01) exhibited significant reduction in the expression

of Akt compared to the rutin (25 mg/kg) treatment group. The expression of Akt in

indomethacin + rutin (2 mg/kg + 25 mg/kg) was non-significant in comparison to the rutin

(25 mg/kg).

3.5.3. Effect of Rutin-GNPs Treatment

The effect of rutin-GNPs on the expression of RANK, RANKL, NF-κB, c-fos, c-jun and Akt

genes is shown in figure 3.21. The expression of all the genes in the arthritic control group

was significantly increased as compared to the normal control animals (p < 0.0001). Both the

treatment groups i.e. indomethacin and rutin-GNPs showed significantly reduced gene

expression of RANK, RANKL, c-fos and c-jun in comparison to the arthritic control group (p

< 0.0001).

The NF-κB and Akt expression in the indomethacin (p < 0.001 and p < 0.0001 for NF-κB and

Akt) treatment group significantly reduced in comparison to the arthritic control group and

similarly the rutin-GNPs treatment also showed significant reduction in gene expression (p <

0.0001 and p < 0.05 for NF-κB and Akt). The statistical analysis showed no significant

difference within the normal control, arthritic control, and treatment groups.

Page 93: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

74

Figure 3.19. The effect of NA-2 on fold change in RANK, RANKL, NF-κB, c-fos, c-

jun, and Akt gene expression. The values are mentioned as mean fold change in gene

expression ± S.E.M. The arthritic control group showed significantly high expression, as

compared to the normal group (#p < 0.0001). A marked suppression in the genes expression

of treatment groups i.e. indomethacin (***p < 0.0001 for RANK, RANKL, c-fos, c-jun, and

Akt, **p < 0.001 for NF-κB) and NA-2 (***p < 0.0001 for RANK, RANKL, NF-κB, c-fos,

and c-jun, *p < 0.01 for Akt) were observed.

Figure 3.20. Effect of rutin on RANK, RANKL, NF-κB, c-fos, c-jun, and Akt gene

expression. The bar graph represents mean fold change in expression ± S.E.M. In

comparison to the normal control group, significantly high gene expressions of the above-

mentioned genes were detected in the arthritic control group (#p < 0.0001). However, the

treatment groups, except rutin (25 mg/kg), exhibited significantly reduced gene

expressions, as compared to the arthritic control rats (*p < 0.01, **p < 0.001, ***p< 0.0001).

#

# ##

#

#

*** ***

**

***

***

********* ***

*** ***

*

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

RANK RANKL NF-κB c-fos c-jun AktFold

Ch

an

ge

in G

ene

Exp

ress

ion

Normal Control Vehicle Control

NA-2 Only Arthritic Control

Arthritic + Vehicle Arthritic + Indomethacin (5 mg/kg)

Arthritic + NA-2 (5 mg/kg)

#

# #

#

#

#

******

**

***

***

****** ***

***

***

***

*

******

***

***

****

0

0.5

1

1.5

2

2.5

3

3.5

4

RANK RANKL NF-kB c-fos c-jun Akt

Fold

Ch

an

ge

in G

ene

Ex

pre

ssio

n

Normal Control Rutin Only

Arthritic Control Arthritic + Indomethacin (5 mg/kg)

Arthritic + Rutin (25 mg/kg) Arthritic + Rutin (50 mg/kg)

Arthritic + Indomethacin + Rutin (2 mg/kg + 25 mg/kg)

Page 94: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

75

Figure 3.21. Expression of RANK, RANKL, NF-κB, c-fos, c-jun, and Akt genes

following rutin-GNPs treatment. The values are mentioned as mean ± S.E.M. Compared

to the normal control rats, fold change in expression of all the genes studied in arthritic

control rats were significantly high (#p < 0.0001). Whereas, the expression of the above-

mentioned genes in rutin-GNPs (12.5 mg/kg) and indomethacin (5 mg/kg) treatment groups

reduced significantly, as compared to the arthritic control group (*p < 0.05, **p < 0.001, ***p < 0.0001).

#

# ##

#

#

*** ***

**

***

***

******

***

***

*** ***

*

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

RANK RANKL NF-κB c-fos c-jun Akt

Fo

ld C

ha

ng

e in

Gen

e E

xp

ress

ion

Normal Control Gold Nanoparticles Only

Arthritic Control Arthritic + Gold Nanoparticles

Arthritic + Indomethacin (5mg/kg) Arthritic + Rutin-GNPs (12.5mg/kg)

Page 95: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

76

3.6. Immunohistochemistry of c-Fos, pAkt and iNOS in Spleen

Tissue

The spleen tissue was isolated from arthritic and non-arthritic rats at the end of in vivo study

and the expression of c-Fos, pAkt and iNOS was determined. The effect of compounds studied

on the expression of above-mentioned genes is shown below. The difference in protein

expression of the arthritic and non-arthritic control groups was non-significant. Therefore, in

microscopic images only normal and arthritic control group is shown and rest of the control

groups are shown in the bar graphs of the compounds.

3.6.1. Effect of NA-2 on c-Fos, pAkt and iNOS Protein Expression

Figure 3.22 shows the mean fluorescent intensity of c-Fos protein in the spleen tissue of

arthritic and non-arthritic animals. Immunohistochemical analysis showed a mild expression

of c-Fos in the normal control group (Fig. 3.23). A significantly high c-Fos expression in the

untreated arthritic control rats was observed in comparison to the normal group (p < 0.0001;

Fig 3.22). In contrast, the c-Fos expression was significantly decreased in the indomethacin

and NA-2 treated groups compared to the arthritic control animals (p < 0.0001). The post hoc

analysis revealed non-significant difference within control and treatment groups.

A baseline expression of pAkt was observed in the normal group (Fig. 3.24). The arthritic

control rats showed significantly high expression of pAkt in comparison to the normal control

group (p < 0.0001, Fig. 3.22). When the NA-2 and indomethacin treatment groups were

analysed for change in expression, a significantly reduced expression of pAkt was observed

compared to the arthritic control group (p < 0.0001). Whereas, change in protein expression

in the treatment groups found to be statistically non-significant.

The iNOS protein expression in spleen tissue samples of all the animals is mentioned in figure

3.25. The statistical analysis showed significantly upregulated iNOS expression in spleen

tissue in the arthritic control animals, compared to the normal control animals (p < 0.0001;

Fig. 3.22). In comparison to the arthritic control rats, NA-2 and indomethacin treatment

groups showed significantly reduced expression of iNOS (p < 0.0001). The comparison of

treatment groups showed non-significant difference.

Page 96: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

77

Figure 3.22. Effect of NA-2 on c-Fos, pAkt and iNOS Protein Expression. Each bar

represents the mean fluorescent intensity of arthritic and non-arthritic rats (n=12

animals/group). A significantly high level of c-Fos expression was observed in the arthritic

control group (#p < 0.0001) as compared with the normal control animals. NA-2 and

indomethacin treatment group showed significant reduction in the expression of c-Fos, pAkt

and iNOS in comparison to the arthritic control group (*p < 0.0001).

# #

#

*

* **

**

0

5

10

15

20

25

C-Fos Akt iNOS

Mea

n F

luo

resc

ence

In

ten

sity

(Arb

itra

ry U

nit

)

Treatment Groups

Normal Control Vehicle Control

NA-2 Only Arthritic Control

Arthritic + Vehicle Arthritic + Indomethacin (5 mg/kg)

Arthritic + NA-2 (5 mg/kg)

Page 97: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

78

Figure 3.23. Effect of NA-2 on immunohistochemistry of c-Fos in spleen tissue.

The figure shows the spleen tissue immunohistochemistry after treatment with NA-2

in arthritic and non-arthritic rats. The panel A and B are normal control and arthritic

control groups. Whereas, C and D are indomethacin and NA-2 treatment group. The

three columns represent DAPI, c-Fos and merged images for each sample.

Significantly high c-Fos expression can be seen in the arthritic control groups in

comparison to the normal groups that showed mild expression. The treatment groups

also showed significantly reduced expression of c-Fos in comparison to the arthritic

control groups. Scale bar corresponds to 25 μm.

Page 98: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

79

Figure 3.24. Effect of NA-2 on Immunohistochemical analysis of pAkt in arthritic

and non-arthritic rats. The normal control group (panel A) has significantly low

pAkt expression compared to the arthritic control (panel B). Whereas, indomethacin

(panel C) and NA-2 (panel D) treatment group also have mild expression similar to

normal group. Scale bar corresponds to 25 μm.

Page 99: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

80

Figure 3.25. Immunohistochemical analysis of iNOS expression after treatment

with NA-2. The figures represent the spleen tissue sections depicting the expression

of iNOS protein in the arthritic and non-arthritic rats after treatment with NA-2

compound. The arthritic control (panel B) represent significantly increased expression

of iNOS compared to the normal control group (panel A). Whereas, indomethacin

(panel C) and NA-2 (panel D) treatment showed significantly reduced expression, as

compared to the arthritic control. Scale bar corresponds to 25 μm.

Page 100: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

81

3.6.2. Effect of Rutin on c-Fos, pAkt, and iNOS Protein Expression

The mean fluorescence intensity of c-Fos protein expression is mentioned in figure 3.26. A

prominent increase in the splenic c-Fos expression was detected in the untreated arthritic

control animals compared to the normal control rats (Fig. 3.27). The statistical analysis

demonstrated a marked increase in the mean fluorescence intensity of the c-Fos expression (p

< 0.0001) in the arthritic control rats. In comparison to arthritic control group, except rutin

(25 mg/kg), all the other treatment groups demonstrated a significantly reduced c-Fos

expression in comparison to the arthritic control group (p < 0.0001). The Bonferroni’s post

hoc test demonstrated that in comparison to 25 mg/kg dose of rutin, the indomethacin, rutin

(50 mg/kg) and indomethacin + rutin (2 mg/kg + 25 mg/kg) groups showed significant

decrease in the c-fos expression (p < 0.0001).

In the arthritic control animals, a marked increase in pAkt protein expression was detected

(Fig. 3.28), when compared with the normal control group and when analysed statistically,

found to be significant (p < 0.0001; Fig. 3.26). The immunohistochemical analysis of the

treatment groups showed that pAkt expression in rutin (25 mg/kg) was statistically non-

significant as compared to the arthritic control rats. Whereas, the rutin (50 mg/kg),

indomethacin (5 mg/kg) and indomethacin + rutin (2 mg/kg + 25 mg/kg) showed significantly

reduced expression (p < 0.0001). The comparison of treatment groups was also done to

determine differences between the treatment groups and significant decrease was observed in

the indomethacin (5 mg/kg, p < 0.001), rutin (50 mg/kg, p < 0.01) and indomethacin + rutin

(2.5 mg/kg + 25 mg/kg, p < 0.05) treatment groups in comparison to rutin (25 mg/kg).

The iNOS expression in the arthritic control exhibited significant increase compared to the

normal control (p < 0.0001, Fig. 3.26 and 3.29). The treatment groups depicted significant

suppression in the iNOS expression (p < 0.0001 for rutin (50 mg/kg), indomethacin (5 mg/kg)

and indomethacin + rutin treatment groups respectively), compared to the arthritic control.

Rutin (25 mg/kg) demonstrated non-significant difference compared to the arthritic control

group. The Post hoc analysis also revealed that all the treatment groups showed significant

reduction in iNOS protein expression compared to rutin (25 mg/kg) treatment group (p <

0.0001). However, non-significant difference was found within the control groups.

Page 101: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

82

Figure 3.26. Effect of rutin on c-Fos, pAkt and iNOS protein expression. The bar graph

shows the mean fluorescent intensity ± S.E.M. of arthritic and non-arthritic rats (n=12

animals/group). The Immunohistochemical analysis of c-fos, pAkt and iNOS proteins

expression in the spleen tissue sample of untreated arthritic animals demonstrated significant

increase, compared to the normal control group (#p < 0.0001). Whereas, all the treatment

groups, except rutin (25 mg/kg) showed significantly reduced expression of afore-mentioned

proteins compared to the arthritic control group (*p < 0.0001).

##

#

*

* ***

** *

*

0

5

10

15

20

25

C-Fos Akt iNOS

Mea

n F

luo

resc

ence

In

ten

sity

(Arb

itra

ry U

nit

)

Treatment Groups

Normal Control Rutin Only

Arthritic Control Arthritic + Indomethacin (5 mg/kg)

Arthritic + Rutin (25 mg/kg) Arthritic + Rutin (50 mg/kg)

Arthritic + Indomethacin + Rutin (2 mg/kg + 25 mg/kg)

Page 102: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

83

Figure 3.27. Immunohistochemistry of c-Fos in rutin treated animals. The

figures represented the c-Fos expression in the spleen tissue of rats. The normal

control (panel A) showed mild c-Fos expression compared to the arthritic control

animals (panel B). Whereas, treatment groups i.e. indomethacin (panel C), rutin

(50 mg/kg, panel E) and indomethacin + rutin (2.5 mg/kg + 25 mg/kg, panel F)

showed significantly reduced c-Fos expression. However, rutin (25 mg/kg, panel

D) showed significantly high expression compared to the normal control. Scale

bar corresponds to 25 μm.

Page 103: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

84

Figure 3.28. Immunohistochemical analysis of pAkt in rutin treated group. The

arthritic control group (panel B) showed high expression of pAkt in the spleen tissue

as compared to the normal control rats (panel A). A prominent suppression in pAkt

expression was observed in the indomethacin (panel C), rutin (50 mg/kg, panel E)

and indomethacin + rutin (2 mg/kg + 25 mg/kg, panel F) treatment groups. The

rutin (25 mg/kg, panel D) group showed non-significant reduction in expression

compared to the arthritic control animals. Scale bar corresponds to 25 μm.

Page 104: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

85

Figure 3.29. Photomicrograph of iNOS expression in rutin treated animals. The figures depicted mild expression of iNOS in the normal control (panel A).

Whereas, high level of iNOS expression was detected in the arthritic control

(panel B), compared to the normal control group. Similarly, rutin (25 mg/kg,

panel D) treatment group showed increased iNOS expression that was

significantly high compared to the rest of the treatment groups. The

indomethacin (panel C), rutin (50 mg/kg, panel E) and indomethacin + rutin

(2.5 mg/kg + 25 mg/kg, panel F) treatment groups showed significantly reduced

iNOS expression in comparison to the arthritic control group. Scale bar

corresponds to 25 μm.

Page 105: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

86

3.6.3. Effect of Rutin-GNPs on c-Fos, pAkt, and iNOS Protein Expression

The immunohistochemical analysis of c-Fos expression in the spleen tissue of the arthritic and

non-arthritic rats is shown in figure 3.30 and 3.31. A prominent increase in the c-Fos

expression was observed in the arthritic control group as compared to the normal control

animals (p < 0.0001). Whereas, both the treatment groups i.e. indomethacin and rutin-GNPs

(12.5 mg/kg) showed low expression level compared to the arthritic control group and this

reduction was statistically significant (p < 0.0001). The Bonferroni’s post hoc test

demonstrated non-significant difference within the control and treated groups.

The protein expression of untreated arthritic control animals exhibited significant

upregulation, compared to the normal control animals (p < 0.0001, Fig. 3.30). The rutin-GNPs

(12.5 mg/kg) and indomethacin (5 mg/kg) treatment groups demonstrated significant

suppression of pAkt protein expression (Fig. 3.32), compared to the arthritic control group (p

< 0.0001). The post hoc analysis showed non-significant difference between the treatment

groups.

The protein expression of iNOS in the spleen tissue of arthritic rats was significantly high

compared to the normal control rats (p < 0.0001, figure 3.30 and 3.33). However, when

treatment groups i.e. indomethacin (5 mg/kg) and rutin-GNPs (12.5 mg/kg) were compared

with the arthritic control group, significant reduction in the protein expression was observed

(p < 0.0001). The difference within the arthritic and non-arthritic control groups and the

treatment groups was non-significant.

Page 106: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

87

Figure 3.30. Effect of rutin-GNPs on c-Fos, pAkt and iNOS expression. The bar graph

represents values as mean ± S.E.M. (n= 12 animals/group). Significantly high c-Fos, pAkt

and iNOS protein expression was observed in the arthritic control animals compared to the

normal control animals (#p < 0.0001). In contrast, reduced expression was detected in the

treatment groups (*p < 0.0001 for both indomethacin and rutin-GNPs treatment group).

##

#

*

* ***

*

0

5

10

15

20

25

C-Fos Akt iNOS

Mea

n F

luo

resc

ence

In

ten

sity

(Arb

itra

ry U

nit

)

Treatment Groups

Normal Control Gold Nanoparticles Only

Arthritic Control Arthritic + Gold Nanoparticles

Arthritic + Indomethacin (5 mg/kg) Arthritic + Rutin-GNPs (12.5 mg/kg)

Page 107: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

88

Figure 3.31. Effect of rutin-GNPs on Immunohistochemical analysis of c-Fos. The

arthritic control (panel B) showed significantly high level of c-Fos expression as

compared to the normal control animals (panel A). Whereas, the treatment groups, i.e.

indomethacin (panel C) and rutin-GNPs (panel D) showed significant reduction in c-

Fos expression. Scale bar corresponds to 25 μm.

Page 108: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

89

Figure 3.32. Immunohistochemistry of pAkt on rutin-GNPs treated rats. The

normal control (panel A), showed low level of pAkt expression in comparison to the

arthritic control (panel B). Whereas, the arthritic groups treated with indomethacin

(panel C) and rutin-GNPs (panel D) showed significantly reduced pAkt expression

compared to the arthritic control group. Scale bar corresponds to 25 μm.

Page 109: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

90

Figure 3.33. Immunohistochemistry of iNOS in spleen tissue of rutin-GNPs

treated group. In comparison to the arthritic control (panel B), the normal control

group (panel A) showed significantly reduced iNOS expression. Similarly, the

indomethacin (panel C) and rutin-GNPs (panel D) treatment group also showed

significantly reduced expression as compared to the arthritic control animals. Scale bar

corresponds to 25 μm.

Page 110: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

91

CHAPTER 4

DISCUSSION

Page 111: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

92

Rheumatoid arthritis is a an inflammatory autoimmune condition (Wong and Lord, 2004).

Continuous decrease in the disease severity is rarely achieved with the use of conventional

synthetic and biological DMARDs and patients often require lifelong treatment with serious

side effects. The precise molecular mechanism involved in the pathogenesis of RA is still

not clear due to which there is a lack of any effective treatment modality of RA. Therefore,

the identification of possible pathogenic mechanism causing the initiation and progression of

RA can result in the development of an effective treatment strategy (Goldring, 2003).

Members of the TNF receptor family and TNF ligand i.e. RANK and RANKL have been

identified and characterized that aid in the understanding of the molecular mechanism that

controls the formation of osteoclasts that are key players in the progression of RA (Lacey,

Timms, Tan, Kelley, Dunstan, Burgess, Elliott, Colombero, Elliott, and Scully, 1998; Myers,

Collier, Minkin, Wang, Holloway, Malakellis, and Nicholson, 1999; Cenci, Weitzmann,

Roggia, Namba, Novack, Woodring, and Pacifici, 2000; Shalhoub, Elliott, Chiu, Manoukian,

Kelley, Hawkins, Davy, Shimamoto, Beck, and Kaufman, 2000; Collin-Osdoby, Rothe,

Anderson, Nelson, Maloney, and Osdoby, 2001). Therefore, in the current study we focused

on the RANK/RANKL pathway to understand the molecular mechanism behind the anti-

arthritic activity of the test compounds studied i.e. NA-2, rutin, indomethacin + rutin and rutin

GNPs in CIA rats.

The test compounds were initially tested in acute carrageenan-induced inflammatory model

and after validating the anti-inflammatory effect, they were further tested in animal model of

arthritis i.e. collagen induced arthritis (CIA) with focus on RANK-RANKL pathway.

Trentham et al reported the use of CIA in rats for the first time after immunisation with type

II collagen (CII), which is one of the main components of articular cartilage. This model has

many similarities to the human arthritic condition in terms of histological, clinical and

immunological characteristics (Trentham, Townes, and Kang, 1977; Rosloniec, Cremer,

Kang, and Myers, 2001). Therefore, it has been used widely in studies involving the

preclinical evaluation of novel drugs for the treatment of RA (Billingham, 1983; Rosloniec,

Cremer, Kang, and Myers, 2001).

To evaluate the effect of NA-2, rutin, indomethacin + rutin and rutin-GNPs treatment on the

progression of CIA, the clinical parameters like change in body weight, paw volume and

Page 112: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

93

arthritic score were examined during the entire study. The increase in paw volume is the result

of synovial infiltration of inflammatory mediators that include reactive oxygen species,

lymphocytes, prostaglandins and cytokines. Studies of inflammatory pain model reported that

the agents causing reduction of paw inflammation also involve in prevention of the

recruitment of cells involve in the inflammatory response towards the arthritic joints that

ultimately results in improvement in the mobility of animals and hence the animals can easily

access food and water (Ghirnikar, Lee, and Eng, 1998; McHugh and McHugh, 2000). Umar

et al reported the suppression of paw score and oedema in CIA rats after treatment with rutin

(Umar, Mishra, Pal, Sajad, Ansari, Ahmad, Katiyar, and Khan, 2012). Guardia et al also

demonstrated the protective effect of rutin on paw oedema and arthrogram score in AIA model

(Guardia, Rotelli, Juarez, and Pelzer, 2001). The observation in the current study is consistent

with the earlier findings. The untreated arthritic rats showed continuous decrease in the

average body weight and increase in paw volume and arthritic score till the end of the study,

whereas, the compounds NA-2, rutin (50 mg/kg), indomethacin + rutin (2 mg/kg + 25 mg/kg),

rutin-GNPs (12.5 mg/kg) and control drug indomethacin (5 mg/kg) treatment groups showed

increment in their body weight and significant reduction in the arthritic score and paw oedema.

Other studies from our research group have also reported the similar findings (Jawed, Shah,

Jamall, and Simjee, 2010; Jawed, Jamall, Shah, Perveen, Hanif, and Simjee, 2014; Perveen,

Hanif, Jawed, Jamall, and Simjee, 2014).

Besides the evaluation of the macroscopic parameters associated with the arthritis progression,

the CIA rats were also examined for the histological changes in the knee joints. We found that

the clinical parameters and histological score of arthritis were correlated well. The features

observed in the arthritic control rats were infiltration of synovial region with leukocytes,

cartilage and bone erosion and synovial cells proliferation. It has been reported in various

studies that the inflammatory response in arthritis leads to destruction of cartilage and

infiltration and proliferation of inflammatory immune cells in the inflamed tissues (Meyer,

Franssen, and Pap, 2006; Hayer, Pundt, Peters, Wunrau, Kühnel, Neugebauer, Strietholt,

Zwerina, Korb, and Penninger, 2009; Karmakar, Kay, and Gravallese, 2010). At the site of

tissue inflammation, activated fibroblast and macrophages leads to the secretion of

inflammatory molecules like prostaglandins, oxidative species, leukotrienes and proteinases

that triggers the process of cartilage destruction (Wieland, Michaelis, Kirschbaum, and

Page 113: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

94

Rudolphi, 2005). The cartilage and bone damage results in the formation of cavity filled with

the synovial fibroblast and inflammatory cells (Ishikawa, Nishigaki, Miyata, Hirayama,

Minoura, Imanishi, Neya, Mizutani, Imamura, and Naritomi, 2005). The motor function in the

RA patients or arthritic animals is hindered due to pain in the joints and changes in the joints

architecture. The treatment strategy that can inhibit or retard the inflammatory response and

joint damage and improve the motor function can be considered therapeutically effective for

RA (Van der Leeden, Steultjens, Dekker, Prins, and Dekker, 2006; Hess, Axmann, Rech,

Finzel, Heindl, Kreitz, Sergeeva, Saake, Garcia, and Kollias, 2011). In our study, we also

observed cartilage damage and erosion in the knee joint tissues of the arthritic control animals

with extensive proliferation of inflammatory cells. However, the histological examination of

the arthritic rats treated with indomethacin and the test compounds (NA-2, rutin-50 mg/kg,

indomethacin + rutin and rutin-GNPs) demonstrated mild synovial infiltration and slight

cartilage damage as compared to the arthritic control rats. These histological analysis correlate

well with macroscopic observations of both arthritic control animals and arthritic animals

treated with the test compounds.

Reactive oxygen and nitrogen species (ROS and RNS) have been known for their role in the

pathophysiology of RA, since long as mediators of cartilage and bone damage. (Panasyuk,

Frati, Ribault, and Mitrovic, 1994; Nakamura, Shibakawa, Tanaka, Kato, and Nishioka, 2003).

The endogenous antioxidant system like GSH and SOD prevent this ROS induced damage. In

arthritic condition this balance of antioxidant and ROS system is disturbed and leads to the

cartilage and bone erosion (Grootveld, Henderson, Farrell, Blake, Parkes, and Haycock, 1991;

Hughes, Reynolds, Brown, Kelley, Thomson, Conn, Jonas, Westfall, Padilla, and Callahan,

2010). Study conducted by Umar et al reported that rutin treatment of arthritic rats leads to

significant suppression of ROS and significant increase in GSH level (Umar, Mishra, Pal,

Sajad, Ansari, Ahmad, Katiyar, and Khan, 2012). Kyung et al also reported that rutin inhibits

osteoclast formation by reducing the elevated ROS level in vitro in RANKL stimulated bone

marrow macrophages (Kyung, Lee, Shin, and Choi, 2008). Our observations were in accord

with these studies, since we have also observed similar findings. The untreated arthritic

animals exhibited significant rise in NO and PO concentration with parallel decrease in the

GSH level compared to the normal control animals. Whereas, the reference drug

indomethacin, NA-2, rutin (50 mg/kg), indomethacin + rutin (2 mg/kg + 25 mg/kg) and rutin-

Page 114: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

95

GNPs (12.5 mg/kg) treatment groups showed significantly reduced NO and PO level with

parallel increase in GSH concentration, as compared to the untreated arthritic group. Several

studies have reported that the inhibitory activity of indomethacin treatment on generation of

ROS is through the inhibition of prostaglandin E2 and COX (Hrabák, Vercruysse, Kahán, and

Vray, 2001; Dalle-Donne, Rossi, Giustarini, Milzani, and Colombo, 2003). These findings

justify the use of indomethacin as a reference drug in the current study.

TNF-α and IL-1β are key players in the progression of RA (Kudo, Fujikawa, Itonaga,

Sabokbar, Torisu, and Athanasou, 2002; Kitaura, Zhou, Kim, Novack, Ross, and Teitelbaum,

2005; Wei, Kitaura, Zhou, Ross, and Teitelbaum, 2005). IL-1β and TNF-α are produced by

various immune cells, mainly activated T cells and macrophages (Jawed, Shah, Jamall, and

Simjee, 2010). TNF-α in combination with RANKL causes upregulation of RANK expression

(Komine, Kukita, Kukita, Ogata, Hotokebuchi, and Kohashi, 2001). It has been reported that

treatment with TNF-α inhibitors reduces inflammation and prevents bone resorption in RA

patients (Scott and Kingsley, 2006). Like TNF-α, studies have also reported protective effects

of IL-1β inhibition on structural damage to bones in animal model of RA (Joosten, Helsen,

Saxne, van de Loo, Heinegård, and van den Berg, 1999). Kyung et al also showed reduction

in TNF-α mRNA expression after treatment of RANKL stimulated bone marrow derived

macrophages with rutin (Kyung, Lee, Shin, and Choi, 2008). Similarly, rutin stabilized silver

nanoparticles also reported significant decrease in TNF-α in animal model of arthritis (Rao,

2015).

Extensive studies have been done on the effect of IL-1β and TNF-α on the progression of

arthritis. All these studies reported elevated level of these cytokines in the arthritic patients

and in vivo models of arthritis (Glenn and Gray, 1965; Ghirnikar, Lee, and Eng, 1998;

McHugh and McHugh, 2000; Coulthard, Pleuvry, Brewster, Wilson, and Macfarlane, 2002;

Laferrère, García-Lorda, Russell, and Pi-Sunyer, 2004; Van der Heijden, Oerlemans, Lems,

Scheper, Dijkmans, and Jansen, 2009). In correlation to the above mentioned findings, we

have also observed high TNF-α and IL-1β concentration in the arthritic animals. Following

treatment with NA-2, rutin (50 mg/kg) and indomethacin + rutin (2 mg/kg + 25 mg/kg), rutin-

GNPs (12.5 mg/kg) and indomethacin, a significantly decreased level of these cytokines was

observed.

Page 115: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

96

Inducible nitric oxide synthase (iNOS) is an isoform of nitric oxide synthase (NOS), that is

expressed in the macrophages after exposure of bacterial endotoxin and pro-inflammatory

cytokine INF-γ, TNF-α and IL-1β (Nathan, 1992; Moncada and Higgs, 1993; McNeill,

Crabtree, Sahgal, Patel, Chuaiphichai, Iqbal, Hale, Greaves, and Channon, 2015). The

inflammatory cytokines induce iNOS production that leads to the increased production of NO

for long period in the affected tissues of RA patients (Aktan, 2004). Tozatto et al reported

increased iNOS protein expression in kidney and cardiac tissue in CIA mice (Palma Zochio

Tozzato, Taipeiro, Spadella, Marabini Filho, de Assis, Carlos, Girol, and Chies, 2015).

Cannon and Tanaka et al also reported upregulation of iNOS mRNA expression in spleen

tissue of CIA and AIA rats as compared to normal rats (Cannon, Openshaw, Hibbs, Hoidal,

Huecksteadt, and Griffiths, 1996; Tanaka, Matsui, Murakami, Ishizuka, Sugiura, Kawashima,

and Sugita, 1998). In light of the above findings, we also observed prominent reduction in

protein expression of iNOS in NA-2, rutin-50 mg/kg, indomethacin + rutin (2 mg/kg + 25

mg/kg), rutin-GNPs (12.5 mg/kg) and indomethacin treatment groups in comparison to the

arthritic control animals that exhibited high level of iNOS expression in spleen tissue.

Extensive studies have been done to determine the role of the RANK/RANKL/OPG signalling

pathway in RA (Neumann, Gay, and Müller‐ Ladner, 2005). RANKL interacts with its

cognate receptor RANK that leads to the further activation of series of downstream signalling

cascade that results in activation of downstream transcription factors NF-κB, c-fos and c-jun

followed by the activation of Akt (Grigoriadis, Wang, Cecchini, Hofstetter, Felix, Fleisch, and

Wagner, 1994; Gravallese, Manning, Tsay, Naito, Pan, Amento, and Goldring, 2000; Lam,

Takeshita, Barker, Kanagawa, Ross, and Teitelbaum, 2000; Ruocco, Maeda, Park, Lawrence,

Hsu, Cao, Schett, Wagner, and Karin, 2005; Wei, Kitaura, Zhou, Ross, and Teitelbaum, 2005).

Animal studies of arthritis have reported that animals genetically deficient in RANK or

RANKL or inhibition of RANKL- RANK interaction leads to protection of cartilage and bone

erosion regardless of the presence of joint inflammation (Simonet, Lacey, Dunstan, Kelley,

Chang, Lüthy, Nguyen, Wooden, Bennett, and Boone, 1997; Kong, Feige, Sarosi, Bolon,

Tafuri, Morony, Capparelli, Li, Elliott, and McCabe, 1999; Pettit, Ji, Von Stechow, Müller,

Goldring, Choi, Benoist, and Gravallese, 2001; Redlich, Hayer, Maier, Dunstan, Tohidast‐

Akrad, Lang, Türk, Pietschmann, Woloszczuk, and Haralambous, 2002; Romas, Sims, Hards,

Page 116: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

97

Lindsay, Quinn, Ryan, Dunstan, Martin, and Gillespie, 2002; Lubberts, van den Bersselaar,

Oppers-Walgreen, Schwarzenberger, Coenen-de Roo, Kolls, Joosten, and van den Berg, 2003;

Li, Schwarz, O'Keefe, Ma, Looney, Ritchlin, Boyce, and Xing, 2004). Tsubaki et al reported

over expression of RANKL mRNA in the spleen and thymus samples of CIA mice (Tsubaki,

Takeda, Kino, Itoh, Imano, Tanabe, Muraoka, Satou, and Nishida, 2015). Wei et al also

reported that the animals with adjuvant induced arthritis showed upregulation of RANKL

(Wei, Tong, Xia, Lu, Chou, Wang, and Dai, 2013).

In the light of above findings, the present study also demonstrated increased mRNA and

protein expression of RANK and RANKL in the spleen samples of CIA rats. However, the

administration of NA-2, rutin (50 mg/kg), indomethacin + rutin (2 mg/kg + 25 mg/kg), rutin-

GNPs (12.5 mg/kg) significantly suppressed the CIA progression by downregulation of

RANK and RANKL that clearly shows the role of RANK/RANKL in the progression of

arthritis.

In addition to RANK and RANKL, some other markers that are downstream mediators of

RANK/RANKL signalling cascade were also studied. In the pathogenesis of RA, NF-κB plays

central role. The role of NF-κB in the regulation of different pro-inflammatory pathways is

well known. NF-κB regulates T and B lymphocytes activation and survival in spleen and

thymus that ultimately leads to the development of arthritis (Brown, Claudio, and Siebenlist,

2008). High activity of NF-κB has been reported in a number of studies involving

experimental arthritis (Tsao, Suzuki, Totsuka, Murata, Takagi, Ohmachi, Fujimura, and

Takata, 1997; Miagkov, Kovalenko, Brown, Didsbury, Cogswell, Stimpson, Baldwin, and

Makarov, 1998; Tak and Firestein, 2001). Activation of the inflammatory mediators involved

in the progression of RA like IL-1β, IL-6, TNF-α and RANKL at the transcriptional level is

dependent on the activation of NF-κB (Tracey, Klareskog, Sasso, Salfeld, and Tak, 2008).

Kyung et al also demonstrated inhibition of NF-κB activation by rutin treatment in RANKL

stimulated bone marrow derived macrophages (Kyung, Lee, Shin, and Choi, 2008). Inhibition

of NF-κB has been reported to suppress the progression of arthritis in various animal models

(Wakamatsu, Nanki, Miyasaka, Umezawa, and Kubota, 2005; Izmailova, Paz, Alencar, Chun,

Schopf, Hepperle, Lane, Harriman, Xu, and Ocain, 2007; Okamoto, Yoshio, Kaneko, and

Yamanaka, 2010). Our findings are also consistent with the above mentioned reports. In the

Page 117: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

98

current study we observed upregulation of NF-κB in the arthritic control rats and the treatment

of NA-2, rutin-50 mg/kg, indomethacin + rutin (2 mg/kg + 25 mg/kg), rutin-GNPs (12.5

mg/kg) and indomethacin significantly suppressed the expression of NF-κB.

Activator protein 1 (AP-1) belongs to the transcription factor family that comprises of homo

and heterodimers of Fos, Jun, ATF and MAF family of proteins (Hai, Liu, Allegretto, Karin,

and Green, 1988; Angel and Karin, 1991). AP-1 is involved in different processes e.g. cell

migration, differentiation, apoptosis, proliferation etc. Beside this, it is also an important

player in inflammatory disorders like psoriasis, asthma and rheumatoid arthritis (Palanki,

2002; Shaulian and Karin, 2002; Hess, Angel, and Schorpp-Kistner, 2004; Wagner and

Eferl, 2005). The activation of P38/MAPKs leads to regulation of downstream mediators

that causes c-fos and c-jun transcriptional activation and finally upregulation of AP-1

complex expression takes place (Eferl and Wagner, 2003). AP-1 activation is an

important step in the process of joint damage (Shiozawa, Shimizu, Tanaka, and Hino,

1997). The transcription factors c-jun and c-fos are key players in the process of bone

resorption and blockade of any of these factors can prevent osteoclastogenesis

(Teitelbaum, 2004; Takayanagi, 2005). It has been reported that blockade of c-fos and c-

jun has prevented the RANKL induced osteoclastogenesis (Ikeda, Nishimura, Matsubara,

Tanaka, Inoue, Reddy, Hata, Yamashita, Hiraga, and Watanabe, 2004; Lee, Jin, Shim, Kim,

Ha, and Lee, 2010). Zhang et al demonstrated upregulated expression of c-fos and c-jun in

RA patients synovial tissue and peripheral blood compared to healthy individuals (Zhang, Yu,

Deng, Lv, Liu, Xiao, Yang, Zhang, and Li, 2015). Hashiramoto et al also reported upregulated

c-Fos protein expression in spleen of CIA rats (Hashiramoto, Yamane, Tsumiyama, Yoshida,

Komai, Yamada, Yamazaki, Doi, Okamura, and Shiozawa, 2010). Liu et al reported increased

mRNA expression of c-jun in LPS induced splenocytes isolated from CFA induced arthritic

rats (Liu, Li, He, Yang, Ruan, and Sun, 2016). Similarly, the present study also showed

upregulation of both c-fos and c-jun at mRNA and protein level in arthritic animals as

compared to the normal healthy rats and this upregulation was suppressed significantly in NA-

2, rutin-50 mg/kg, indomethacin + rutin (2 mg/kg + 25 mg/kg), rutin-GNPs (12.5 mg/kg), and

indomethacin treated groups.

Page 118: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

99

Another downstream mediator of inflammatory cascade is Akt which is also known as protein

kinase B, belong to serine/threonine kinase family of proteins (King, Kobayashi, Cejas, Kim,

Yoon, Kim, Chiffoleau, Hickman, Walsh, and Turka, 2006). It is expressed mainly in the cells

of the innate immunity e.g. macrophages, neutrophils and dendritic cells. It is a key player in

the activation of different signalling cascades involved in apoptosis, cell growth etc. (Huang

and Lin, 2007; Fischer, Jacoby, Pape, Ward, Kuwertz-Broeking, Renken, Nizze, Querfeld,

Rudolph, and Mueller-Wiefel, 2009; Emamian, 2012). Studies have identified its role in

autoimmune diseases. AKT signalling pathway is involved in the regulation of inflammation.

Various Studies have reported the role of PI3K/AKT pathway in the inflammatory disorders

such as multiple sclerosis, asthma, psoriasis, RA and atherosclerosis (Bozinovski, Jones,

Vlahos, Hamilton, and Anderson, 2002; Chang, Sukhova, Libby, Schvartz, Lichtenstein,

Field, Kennedy, Madhavarapu, Luo, and Wu, 2007; Mortaz, Chiu, Lin, Chen, Huang, Tong,

Tzeng, Lee, Hsu, and Tang, 2009; Lazar, Koenderman, Kraneveld, Nijkamp, and Folkerts,

2009; Ma, Dela Cruz, Hartl, Kang, Takyar, Homer, Lee, and Elias, 2011; Lai, Li, Li,

Muehleisen, Radek, Park, Jiang, Li, Lei, and Quan, 2012). Tsubaki et al reported increased

activation of Akt in CIA mice (Tsubaki, Takeda, Kino, Itoh, Imano, Tanabe, Muraoka, Satou,

and Nishida, 2015). Wei et al also reported significant increase in phospho-Akt expression in

AIA rats (Wei, Tong, Xia, Lu, Chou, Wang, and Dai, 2013). In the current study, we also

found increased expression of pAkt in arthritic rats that decreased significantly, when the

animals were given NA-2, rutin-50 mg/kg, indomethacin + rutin (2 mg/kg + 25 mg/kg), rutin-

GNPs (12.5 mg/kg) and indomethacin treatment.

Page 119: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

100

CONCLUSION

The present study has demonstrated that N-(2-hydroxyl phenyl) acetamide (NA-2), rutin,

indomethacin + rutin and rutin GNPs not only prevented the clinical signs of arthritis, ROS

and pro-inflammatory cytokines production in CIA model but for the first time demonstrated

that these compounds have significantly downregulated RANKL and its receptor RANK

expression and successfully suppressed the expression of other pro-inflammatory mediators

downstream of RANKL signalling cascade such as c-Fos, c-Jun, NF-κB and Akt in CIA

model. The compounds significantly reduced the arthritic score, paw oedema and the

inflammatory markers level in CIA rats. Our data suggest that expression of mediators of

RANKL pathway, along with ROS and circulating pro-inflammatory cytokines, can be used

to monitor the efficacy of treatment in animal model of RA. In our study, the expression of all

these markers of inflammation correlate well with the clinical symptoms in the untreated

arthritic group and the test compounds treatment successfully downregulated the level of these

markers. Thus, based on our data analysis, we found that RANKL and its receptor RANK

may be used as markers of disease induction. In this regard activation of RANKL can be a

useful tool after nociceptive stimulation to identify the effectiveness of different analgesics

used in the RA treatment. Taken together, it can be concluded that NA-2, rutin, and rutin-

GNPs might be potential candidates for RA treatment.

Page 120: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

101

FUTURE IMPLICATIONS

In the current study, NA-2, rutin and rutin-GNPs showed promising anti-arthritic and anti-

inflammatory activity in CIA model of rat. We investigated the potential role of these

compounds in the regulation of RANK-RANKL pathway and to some extent we were able to

identify the key mediators downstream of RANKL signalling cascade but in the progression

of RA crosstalk of multiple pathways is involved and to get the complete information of RA

progression, other signalling pathways also need to be investigated. Therefore, the future

targets will be:

1. To study IL-6/STAT3 signalling pathway. It is constitutively activated in monocytes and

T lymphocytes circulating T cells of RA patients. Limited information is available on the

role of STAT3 pathway in the progression of RA. Investigation of effect of NA-2, rutin

and its gold nanoparticles on modulation of STAT3 pathway will be beneficial for the

development of targeted therapies for RA patients.

2. TNF-α and IL-1β production stimulates the expression of matrix metalloproteinases in RA

patients. These enzymes are involved in the destruction of extracellular matrix. Research

is going on to develop potent inhibitors of MMPs to prevent the cartilage and tissue

damage in RA patients. Therefore, future studies can include the determination of NA-2,

rutin and its gold nanoparticles effect on the expression of MMPs.

3. To determine the role of these compounds in the modulation of MEK/ERK signalling

pathway in CIA rats. Activation of MEK/ERK signalling cascade is responsible for the

production of various cytokines like IL-6, IL-1β, TNF-α and RANKL that are main players

responsible for the inflammatory response in RA.

4. Rheumatoid arthritis is a systemic disease and it affects multiple systems of human body.

Therefore, besides immune system which is mainly involved in autoimmune response

regulation, the role of studied compounds in modulation of RANKL pathway can also be

determined in other organ systems like bone, cartilage and cardiovascular system.

Page 121: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

102

REFERENCES

Page 122: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

103

Afonso, V., Champy, R., Mitrovic, D., Collin, P., and Lomri, A. (2007). Reactive oxygen

species and superoxide dismutases: role in joint diseases. Joint Bone Spine, 74, 324-329.

Aktan, F. (2004). iNOS-mediated nitric oxide production and its regulation. Life Sciences, 75,

639-653.

Anderson, D. M., Maraskovsky, E., Billingsley, W. L., Dougall, W. C., Tometsko, M. E.,

Roux, E. R., Teepe, M. C., DuBose, R. F., Cosman, D., and Galibert, L. (1997). A homologue

of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature,

390, 175-179.

Andersson, M., and Holmdahl, R. (1990). Analysis of type II collagen‐ reactive T cells in the

mouse I. Different regulation of autoreactive vs. non‐ autoreactive anti‐ type II collagen T

cells in the DBA/1 mouse. European Journal of Immunology, 20, 1061-1066.

Angel, P., and Karin, M. (1991). The role of Jun, Fos and the AP-1 complex in cell-

proliferation and transformation. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer,

1072, 129-157.

Auger, I., and Roudier, J. (1997). A function for the QKRAA amino acid motif: mediating

binding of DnaJ to DnaK. Implications for the association of rheumatoid arthritis with HLA-

DR4. Journal of Clinical Investigation, 99, 1818.

Bakharevski, O., Stein-Oakley, A. N., Thomson, N. M., and Ryan, P. F. J. (1998). Collagen-

induced arthritis in rats: contrasting effect of subcutaneous versus intradermal inoculation of

type II collagen. Jounal of Rheumatology, 25, 1945–52.

Bedard, K., and Krause, K. H. (2007). The NOX family of ROS-generating NADPH oxidases:

physiology and pathophysiology. Physiological Reviews, 87, 245-313.

Beltrán, B., Mathur, A., Duchen, M. R., Erusalimsky, J. D., and Moncada, S. (2000). The

effect of nitric oxide on cell respiration: A key to understanding its role in cell survival or

death. Proceedings of the National Academy of Sciences, 97, 14602-14607.

Bhattacharyya, A., Chattopadhyay, R., Mitra, S., and Crowe, S. E. (2014). Oxidative stress:

an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiological

Reviews, 94, 329-354.

Billingham, M. (1983). Models of arthritis and the search for anti-arthritic drugs.

Pharmacology & Therapeutics, 21, 389-428.

Bishnoi, M., Chopra, K., and Kulkarni, S. K. (2007). Protective effect of rutin, a polyphenolic

flavonoid against haloperidol‐ induced orofacial dyskinesia and associated behavioural,

biochemical and neurochemical changes. Fundamental & Clinical Pharmacology, 21, 521-

529.

Page 123: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

104

Blair, I. A. (2006). Endogenous glutathione adducts. Current Drug Metabolism, 7, 853-872.

Bozinovski, S., Jones, J. E., Vlahos, R., Hamilton, J. A., and Anderson, G. P. (2002).

Granulocyte/macrophage-colony-stimulating factor (GM-CSF) regulates lung innate

immunity to lipopolysaccharide through Akt/Erk activation of NFκB and AP-1 in vivo.

Journal of Biological Chemistry, 277, 42808-42814.

Brennan, F. M., and McInnes, I. B. (2008). Evidence that cytokines play a role in rheumatoid

arthritis. The Journal of Clinical Investigation, 118, 3537-3545.

Brennan, F., Jackson, A., Chantry, D., Maini, R., and Feldmann, M. (1989). Inhibitory effect

of TNFα antibodies on synovial cell interleukin-1 production in rheumatoid arthritis. The

Lancet, 334, 244-247.

Brown, G. C. (1999). Nitric oxide and mitochondrial respiration. Biochimica et Biophysica

Acta (BBA)-Bioenergetics, 1411, 351-369.

Brown, K. D., Claudio, E., and Siebenlist, U. (2008). The roles of the classical and alternative

nuclear factor-kappaB pathways: potential implications for autoimmunity and rheumatoid

arthritis. Arthritis Research & Therapy, 10, 1-14.

Cannon, G. W., Openshaw, S. J., Hibbs, J. B., Hoidal, J. R., Huecksteadt, T. P., and Griffiths,

M. M. (1996). Nitric oxide production during adjuvant‐ induced and collagen‐ induced

arthritis. Arthritis & Rheumatism, 39, 1677-1684.

Cenci, S., Weitzmann, M. N., Roggia, C., Namba, N., Novack, D., Woodring, J., and Pacifici,

R. (2000). Estrogen deficiency induces bone loss by enhancing T-cell production of TNF-α.

The Journal of Clinical Investigation, 106, 1229-1237.

Chang, J. D., Sukhova, G. K., Libby, P., Schvartz, E., Lichtenstein, A. H., Field, S. J.,

Kennedy, C., Madhavarapu, S., Luo, J., and Wu, D. (2007). Deletion of the phosphoinositide

3-kinase p110γ gene attenuates murine atherosclerosis. Proceedings of the National Academy

of Sciences, 104, 8077-8082.

Charatcharoenwitthaya, N., Khosla, S., Atkinson, E. J., McCready, L. K., and Riggs, B. L.

(2007). Effect of Blockade of TNF‐ α and Interleukin‐ 1 Action on Bone Resorption in Early

Postmenopausal Women. Journal of Bone and Mineral Research, 22, 724-729.

Chiavaroli, V., Giannini, C., De Marco, S., Chiarelli, F., and Mohn, A. (2011). Unbalanced

oxidant–antioxidant status and its effects in pediatric diseases. Redox Report, 16, 101-107.

Chiu, Y.-C., Lin, C.-Y., Chen, C.-P., Huang, K.-C., Tong, K.-M., Tzeng, C.-Y., Lee, T.-S.,

Hsu, H.-C., and Tang, C.-H. (2009). Peptidoglycan enhances IL-6 production in human

synovial fibroblasts via TLR2 receptor, focal adhesion kinase, Akt, and AP-1-dependent

pathway. The Journal of Immunology, 183, 2785-2792.

Chizzolini, C., Dayer, J.-M., and Miossec, P. (2009). Cytokines in chronic rheumatic diseases:

is everything lack of homeostatic balance? Arthritis Research & Therapy, 11, 246.

Page 124: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

105

Choi, J. W. (2003). Nitric oxide production is increased in patients with rheumatoid arthritis

but does not correlate with laboratory parameters of disease activity. Clinica Chimica Acta,

336, 83-87.

Chomczynski, P., and Sacchi, N. (1987). Single-step method of RNA isolation by acid

guanidinium thiocyanate-phenol-chloroform extraction. Analytical Biochemistry, 162, 156-

159.

Cimen, M., Çimen, Ö., Kacmaz, M., Öztürk, H., Yorgancioğlu, R., and Durak, I. (2000).

Oxidant/antioxidant status of the erythrocytes from patients with rheumatoid arthritis. Clinical

Rheumatology, 19, 275-277.

Clark, M. A., Plank, L. D., Connolly, A. B., Streat, S. J., Hill, A. A., Gupta, R., Monk, D. N.,

Shenkin, A., and Hill, G. L. (1998). Effect of a chimeric antibody to tumor necrosis factor-

alpha on cytokine and physiologic responses in patients with severe sepsis-a randomized,

clinical trial. Critical Care Medicine, 26, 1650-1659.

Cohen, J., and Carlet, J. (1996). INTERSEPT: an international, multicenter, placebo-

controlled trial of monoclonal antibody to human tumor necrosis factor-alpha in patients with

sepsis. Critical Care Medicine, 24, 1431-1440.

Collard, C. D., Agah, A., and Stahl, G. L. (1998). Complement activation following

reoxygenation of hypoxic human endothelial cells: role of intracellular reactive oxygen

species, NF-κB and new protein synthesis. Immunopharmacology, 39, 39-50.

Collin-Osdoby, P., Rothe, L., Anderson, F., Nelson, M., Maloney, W., and Osdoby, P. (2001).

RANKL and OPG expression by human microvascular endothelial cells, regulation by

inflammatory cytokines, and role in human osteoclastogenesis. Journal of Biological

chemistry, 276, 20659-20672.

Cooper, E. R. (2010). Nanoparticles: a personal experience for formulating poorly water

soluble drugs. Journal of Controlled Release, 141, 300-302.

Cope, A. P. (2008). T cells in rheumatoid arthritis. Arthritis Research & Therapy, 10, 1-10.

doi:10.1186/ar2412

Cornish, A. L., Campbell, I. K., McKenzie, B. S., Chatfield, S., and Wicks, I. P. (2009). G-

CSF and GM-CSF as therapeutic targets in rheumatoid arthritis. Nature Reviews

Rheumatology, 5, 554-559.

Coulthard, P., Pleuvry, B. J., Brewster, M., Wilson, K. L., and Macfarlane, T. V. (2002). Gait

analysis as an objective measure in a chronic pain model. Journal of Neuroscience Methods,

116, 197-213.

Cronstein, B. N., Eberle, M. A., Gruber, H. E., and Levin, R. I. (1991). Methotrexate inhibits

neutrophil function by stimulating adenosine release from connective tissue cells.

Proceedings of the National Academy of Sciences, 88, 2441-2445.

Page 125: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

106

Cronstein, B. N., Naime, D., and Ostad, E. (1993). The antiinflammatory mechanism of

methotrexate. Increased adenosine release at inflamed sites diminishes leukocyte

accumulation in an in vivo model of inflammation. Journal of Clinical Investigation, 92, 2675.

Cronstein, B. N., Naime, D., and Ostad, E. (1995). The antiinflammatory effects of

methotrexate are mediated by adenosine Purine and Pyrimidine Metabolism in Man VIII (pp.

411-416). New York: Springer.

Crotti, T. N., Smith, M. D., Findlay, D., Zreiqat, H., Ahern, M. J., Weedon, H., Hatzinikolous,

G., Capone, M., Holding, C., and Haynes, D. R. (2004). Factors regulating osteoclast

formation in human tissues adjacent to peri-implant bone loss: Expression of receptor

activator NFκB, RANK ligand and osteoprotegerin. Biomaterials, 25, 565-573.

Crotti, T., Smith, M. D., Hirsch, R., Soukoulis, S., Weedon, H., Capone, M., Ahern, M. J.,

and Haynes, D. (2003). Receptor activator NF κB ligand (RANKL) and osteoprotegerin

(OPG) protein expression in periodontitis. Journal of Periodontal Research, 38, 380-387.

Crotti, T., Smith, M., Weedon, H., Ahern, M., Findlay, D., Kraan, M., Tak, P., and Haynes,

D. (2002). Receptor activator NF-κB ligand (RANKL) expression in synovial tissue from

patients with rheumatoid arthritis, spondyloarthropathy, osteoarthritis, and from normal

patients: semiquantitative and quantitative analysis. Annals of the Rheumatic Diseases, 61,

1047-1054.

Dalle-Donne, I., Rossi, R., Giustarini, D., Milzani, A., and Colombo, R. (2003). Protein

carbonyl groups as biomarkers of oxidative stress. Clinica Chimica Acta, 329, 23-38.

Das Neves, J. (2014). Co-association of methotrexate and SPIONs into anti-CD64 antibody-

conjugated PLGA nanoparticles for theranostic application. International Journal of

Nanomedicine, 9, 4911-4922.

Datta, S., Kundu, S., Ghosh, P., De, S., Ghosh, A., and Chatterjee, M. (2014). Correlation of

oxidant status with oxidative tissue damage in patients with rheumatoid arthritis. Clinical

Rheumatology, 33, 1557-1564.

De Rycke, L., Peene, I., Hoffman, I., Kruithof, E., Union, A., Meheus, L., Lebeer, K., Wyns,

B., Vincent, C., and Mielants, H. (2004). Rheumatoid factor and anticitrullinated protein

antibodies in rheumatoid arthritis: diagnostic value, associations with radiological progression

rate, and extra-articular manifestations. Annals of the Rheumatic Diseases, 63, 1587-1593.

Deavall, D. G., Martin, E. A., Horner, J. M., and Roberts, R. (2012). Drug-induced oxidative

stress and toxicity. Journal of Toxicology, 2012, 1-13. doi:10.1155/2012/645460.

Di Carlo, G., Mascolo, N., Izzo, A. A., and Capasso, F. (1999). Flavonoids: old and new

aspects of a class of natural therapeutic drugs. Life sciences, 65, 337-353.

Doran, M. F., Crowson, C. S., Pond, G. R., O'Fallon, W. M., and Gabriel, S. E. (2002).

Frequency of infection in patients with rheumatoid arthritis compared with controls: a

population‐ based study. Arthritis & Rheumatology, 46, 2287-2293.

Page 126: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

107

Dröge, W., Schulze-Osthoff, K., Mihm, S., Galter, D., Schenk, H. e., Eck, H., Roth, S., and

Gmünder, H. (1994). Functions of glutathione and glutathione disulfide in immunology and

immunopathology. The FASEB Journal, 8, 1131-1138.

Eferl, R., and Wagner, E. F. (2003). AP-1: a double-edged sword in tumorigenesis. Nature

Reviews Cancer, 3, 859-868.

Emamian, E. S. (2012). AKT/GSK3 signaling pathway and schizophrenia. Frontiers in

Molecular Neuroscience, 5, 1-12. doi: 10.3389/fnmol.2012.00033.

Farrell, A. J., Blake, D. R., Palmer, R., and Moncada, S. (1992). Increased concentrations of

nitrite in synovial fluid and serum samples suggest increased nitric oxide synthesis in

rheumatic diseases. Annals of the Rheumatic Diseases, 51, 1219-1222.

Fata, J. E., Kong, Y.-Y., Li, J., Sasaki, T., Irie-Sasaki, J., Moorehead, R. A., Elliott, R., Scully,

S., Voura, E. B., and Lacey, D. L. (2000). The osteoclast differentiation factor

osteoprotegerin-ligand is essential for mammary gland development. Cell, 103, 41-50.

Fay, J., Varoga, D., Wruck, C. J., Kurz, B., Goldring, M. B., and Pufe, T. (2006). Reactive

oxygen species induce expression of vascular endothelial growth factor in chondrocytes and

human articular cartilage explants. Arthritis Research & Therapy, 8, R189. doi:

10.1186/ar2102.

Feldmann, M., Brennan, F. M., and Maini, R. N. (1996). Role of cytokines in rheumatoid

arthritis. Annual Review of Immunology, 14, 397-440.

Filippin, L. I., Vercelino, R., Marroni, N., and Xavier, R. M. (2008). Redox signalling and the

inflammatory response in rheumatoid arthritis. Clinical & Experimental Immunology, 152,

415-422.

Firestein, G. S., Budd, R., Gabriel, S. E., McInnes, I. B., and O'Dell, J. R. (2012). Kelley's

textbook of rheumatology: Elsevier Health Sciences.

Fischer, D.-C., Jacoby, U., Pape, L., Ward, C. J., Kuwertz-Broeking, E., Renken, C., Nizze,

H., Querfeld, U., Rudolph, B., and Mueller-Wiefel, D. E. (2009). Activation of the

AKT/mTOR pathway in autosomal recessive polycystic kidney disease (ARPKD).

Nephrology Dialysis Transplantation, 24, 1819-1827.

Fisher Jr, C. J., Agosti, J. M., Opal, S. M., Lowry, S. F., Balk, R. A., Sadoff, J. C., Abraham,

E., Schein, R. M., and Benjamin, E. (1996). Treatment of septic shock with the tumor necrosis

factor receptor: Fc fusion protein. New England Journal of Medicine, 334, 1697-1702.

Formica, J., and Regelson, W. (1995). Review of the biology of quercetin and related

bioflavonoids. Food and Chemical Toxicology, 33, 1061-1080.

Furchgott, R. F., and Zawadzki, J. V. (1980). The obligatory role of endothelial cells in the

relaxation of arterial smooth muscle by acetylcholine. Nature, 288, 373-376.

Page 127: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

108

Gabbita, S. P., Robinson, K. A., Stewart, C. A., Floyd, R. A., and Hensley, K. (2000). Redox

regulatory mechanisms of cellular signal transduction. Archives of Biochemistry and

Biophysics, 376, 1-13.

Gallelli, L., Galasso, O., Falcone, D., Southworth, S., Greco, M., Ventura, V., Romualdi, P.,

Corigliano, A., Terracciano, R., and Savino, R. (2013). The effects of nonsteroidal anti-

inflammatory drugs on clinical outcomes, synovial fluid cytokine concentration and signal

transduction pathways in knee osteoarthritis. A randomized open label trial. Osteoarthritis

and Cartilage, 21, 1400-1408.

Ghirnikar, R. S., Lee, Y. L., and Eng, L. F. (1998). Inflammation in traumatic brain injury:

role of cytokines and chemokines. Neurochemical Research, 23, 329-340.

Glenn, E. M., and Gray, J. (1965). Adjuvant-induced polyarthritis in rats: biologic and

histologic background. American Journal of Veterinary Research, 26, 1180-1194.

Goldring, S. (2003). Inflammatory mediators as essential elements in bone remodeling.

Calcified Tissue International, 73, 97-100.

Goldring, S. R. (2003). Osteoporosis and rheumatic diseases. Primer on the Metabolic Bone

Diseases and Disorders of Mineral Metabolism, 299-301.

Gravallese, E. M., Manning, C., Tsay, A., Naito, A., Pan, C., Amento, E., and Goldring, S. R.

(2000). Synovial tissue in rheumatoid arthritis is a source of osteoclast differentiation factor.

Arthritis & Rheumatism, 43, 250.

Gregersen, P. K., Silver, J., and Winchester, R. J. (1987). The shared epitope hypothesis. An

approach to understanding the molecular genetics of susceptibility to rheumatoid arthritis.

Arthritis & Rheumatism, 30, 1205-1213.

Gremillion, R. B., and Van Vollenhoven, R. F. (1998). Rheumatoid arthritis: designing and

implementing a treatment plan. Postgraduate Medicine, 103, 103-123.

Grigoriadis, A. E., Wang, Z.-Q., Cecchini, M. G., Hofstetter, W., Felix, R., Fleisch, H. A.,

and Wagner, E. F. (1994). c-Fos: a key regulator of osteoclast-macrophage lineage

determination and bone remodeling. Science, 266, 443-449.

Grootveld, M., Henderson, E. B., Farrell, A., Blake, D. R., Parkes, H., and Haycock, P. (1991).

Oxidative damage to hyaluronate and glucose in synovial fluid during exercise of the inflamed

rheumatoid joint. Detection of abnormal low-molecular-mass metabolites by proton-nmr

spectroscopy. Biochemical Journal, 273, 459-467.

Guardia, T., Rotelli, A. E., Juarez, A. O., and Pelzer, L. E. (2001). Anti-inflammatory

properties of plant flavonoids. Effects of rutin, quercetin and hesperidin on adjuvant arthritis

in rat. Il farmaco, 56, 683-687.

Hadjigogos, K. (2003). The role of free radicals in the pathogenesis of rheumatoid arthritis.

Panminerva Medica, 45, 7-13.

Page 128: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

109

Hafström, I., Ringertz, B., Spångberg, A., Von Zweigbergk, L., Brannemark, S., Nylander, I.,

Rönnelid, J., Laasonen, L., and Klareskog, L. (2001). A vegan diet free of gluten improves

the signs and symptoms of rheumatoid arthritis: the effects on arthritis correlate with a

reduction in antibodies to food antigens. Rheumatology, 40, 1175-1179.

Hai, T., Liu, F., Allegretto, E., Karin, M., and Green, M. (1988). A family of immunologically

related transcription factors that includes multiple forms of ATF and AP-1. Genes &

Development, 2, 1216-1226.

Halliwell, B., and Cross, C. E. (1994). Oxygen-derived species: their relation to human disease

and environmental stress. Environmental Health Perspectives, 102, 5.

Haringman, J. J., Gerlag, D. M., Zwinderman, A. H., Smeets, T. J., Kraan, M. C., Baeten, D.,

McInnes, I. B., Bresnihan, B., and Tak, P. P. (2005). Synovial tissue macrophages: a sensitive

biomarker for response to treatment in patients with rheumatoid arthritis. Annals of the

Rheumatic Diseases, 64, 834-838.

Hashiramoto, A., Yamane, T., Tsumiyama, K., Yoshida, K., Komai, K., Yamada, H.,

Yamazaki, F., Doi, M., Okamura, H., and Shiozawa, S. (2010). Mammalian clock gene

Cryptochrome regulates arthritis via proinflammatory cytokine TNF-α. The Journal of

Immunology, 184, 1560-1565.

Hayer, S., Pundt, N., Peters, M. A., Wunrau, C., Kühnel, I., Neugebauer, K., Strietholt, S.,

Zwerina, J., Korb, A., and Penninger, J. (2009). PI3Kγ regulates cartilage damage in chronic

inflammatory arthritis. The FASEB Journal, 23, 4288-4298.

Haynes, D., Crotti, T., Potter, A., Loric, M., Atkins, G., Howie, D., and Findlay, D. (2001).

The osteoclastogenic molecules RANKL and RANK are associated with periprosthetic

osteolysis. Bone & Joint Journal, 83, 902-911.

Haynes, D., Crotti, T., Weedon, H., Slavotinek, J., Au, V., Coleman, M., Roberts‐ Thomson,

P. J., Ahern, M., and Smith, M. D. (2008). Modulation of RANKL and osteoprotegerin

expression in synovial tissue from patients with rheumatoid arthritis in response to disease‐modifying antirheumatic drug treatment and correlation with radiologic outcome. Arthritis

Care & Research, 59, 911-920.

Henrotin, Y., Bruckner, P., and Pujol, J. P. (2003). The role of reactive oxygen species in

homeostasis and degradation of cartilage. Osteoarthritis and Cartilage, 11, 747-755.

Hensley, K., Robinson, K. A., Gabbita, S. P., Salsman, S., and Floyd, R. A. (2000). Reactive

oxygen species, cell signaling, and cell injury. Free Radical Biology and Medicine, 28, 1456-

1462.

Hess, A., Axmann, R., Rech, J., Finzel, S., Heindl, C., Kreitz, S., Sergeeva, M., Saake, M.,

Garcia, M., and Kollias, G. (2011). Blockade of TNF-α rapidly inhibits pain responses in the

central nervous system. Proceedings of the National Academy of Sciences, 108, 3731-3736.

Hess, J., Angel, P., and Schorpp-Kistner, M. (2004). AP-1 subunits: quarrel and harmony

among siblings. Journal of Cell Science, 117, 5965-5973.

Page 129: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

110

Hetland, M. L., Stengaard-Pedersen, K., Junker, P., Lottenburger, T., Hansen, I., Andersen,

L. S., Tarp, U., Svendsen, A., Pedersen, J. K., and Skjødt, H. (2008). Aggressive combination

therapy with intra-articular glucocorticoid injections and conventional disease-modifying

anti-rheumatic drugs in early rheumatoid arthritis: second-year clinical and radiographic

results from the CIMESTRA study. Annals of the Rheumatic Diseases, 67, 815-822.

Hofbauer, L. C., Khosla, S., Dunstan, C. R., Lacey, D. L., Boyle, W. J., and Riggs, B. L.

(2000). The roles of osteoprotegerin and osteoprotegerin ligand in the paracrine regulation of

bone resorption. Journal of Bone and Mineral Research, 15, 2-12.

Holding, C. A., Findlay, D. M., Stamenkov, R., Neale, S. D., Lucas, H., Dharmapatni, A.,

Callary, S. A., Shrestha, K. R., Atkins, G. J., and Howie, D. W. (2006). The correlation of

RANK, RANKL and TNFα expression with bone loss volume and polyethylene wear debris

around hip implants. Biomaterials, 27, 5212-5219.

Hollman, P., and Katan, M. (1997). Absorption, metabolism and health effects of dietary

flavonoids in man. Biomedicine & Pharmacotherapy, 51, 305-310.

Holmdahl, R., Andersson, M. E., Goldschmidt, T. J., Jansson, L., Karlsson, M., Malmstrom,

V., and Mo, J. (1989). Collagen induced arthritis as an experimental model for rheumatoid

arthritis. Apmis, 97, 575-584.

Hørslev-Petersen, K., Hetland, M. L., Junker, P., Pødenphant, J., Ellingsen, T., Ahlquist, P.,

Lindegaard, H., Linauskas, A., Schlemmer, A., and Dam, M. Y. (2013). Adalimumab added

to a treat-to-target strategy with methotrexate and intra-articular triamcinolone in early

rheumatoid arthritis increased remission rates, function and quality of life. The OPERA Study:

an investigator-initiated, randomised, double-blind, parallel-group, placebo-controlled trial.

Annals of the Rheumatic Diseases, 73, 654-61. doi: 10.1136/annrheumdis-2012-202735.

Hrabák, A., Vercruysse, V., Kahán, I. L., and Vray, B. (2001). Indomethacin prevents the

induction of inducible nitric oxide synthase in murine peritoneal macrophages and decreases

their nitric oxide production. Life Sciences, 68, 1923-1930.

Huang, J., and Lin, T. (2007). mTOR signal pathway and its inhibitors in antitumor therapy:

a review. Chinese Journal of Cancer, 26, 1397-1403.

Hughes, L. B., Reynolds, R. J., Brown, E. E., Kelley, J. M., Thomson, B., Conn, D. L., Jonas,

B. L., Westfall, A. O., Padilla, M. A., and Callahan, L. F. (2010). Most common single‐nucleotide polymorphisms associated with rheumatoid arthritis in persons of European

ancestry confer risk of rheumatoid arthritis in African Americans. Arthritis & Rheumatism,

62, 3547-3553.

Ikeda, F., Nishimura, R., Matsubara, T., Tanaka, S., Inoue, J.-i., Reddy, S. V., Hata, K.,

Yamashita, K., Hiraga, T., and Watanabe, T. (2004). Critical roles of c-Jun signaling in

regulation of NFAT family and RANKL-regulated osteoclast differentiation. The Journal of

Clinical Investigation, 114, 475-484.

Page 130: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

111

Inglis, J. J., Notley, C. A., Essex, D., Wilson, A. W., Feldmann, M., Anand, P., and Williams,

R. (2007). Collagen‐ induced arthritis as a model of hyperalgesia: functional and cellular

analysis of the analgesic actions of tumor necrosis factor blockade. Arthritis & Rheumatology,

56, 4015-4023.

Irani, K. (2000). Oxidant signaling in vascular cell growth, death, and survival. Circulation

Research, 87, 179-183.

Isaacs, J., Manna, V., Rapson, N., Bulpitt, K., Hazleman, B., Matteson, E. L., Clair, E. S.,

Schnitzer, T., and Johnston, J. (1996). CAMPATH-1H in rheumatoid arthritis—an

intravenous dose-ranging study. Rheumatology, 35, 231-240.

Ishikawa, T., Nishigaki, F., Miyata, S., Hirayama, Y., Minoura, K., Imanishi, J., Neya, M.,

Mizutani, T., Imamura, Y., and Naritomi, Y. (2005). Prevention of progressive joint

destruction in collagen‐ induced arthritis in rats by a novel matrix metalloproteinase inhibitor,

FR255031. British Journal of Pharmacology, 144, 133-143.

Iuchi, T., Akaike, M., Mitsui, T., Ohshima, Y., Shintani, Y., Azuma, H., and Matsumoto, T.

(2003). Glucocorticoid excess induces superoxide production in vascular endothelial cells and

elicits vascular endothelial dysfunction. Circulation Research, 92, 81-87.

Izmailova, E. S., Paz, N., Alencar, H., Chun, M., Schopf, L., Hepperle, M., Lane, J. H.,

Harriman, G., Xu, Y., and Ocain, T. (2007). Use of molecular imaging to quantify response

to IKK‐ 2 inhibitor treatment in murine arthritis. Arthritis & Rheumatism, 56, 117-128.

Jacobsson, L. T., Hanson, R. L., Knowler, W. C., Pillemer, S., Pettitt, D. J., Mccance, D. R.,

and Bennett, P. H. (1994). Decreasing incidence and prevalence of rheumatoid arthritis in

pima indians over a twenty‐ five year period. Arthritis & Rheumatology, 37, 1158-1165.

Jawed, H., Jamall, S., Shah, S. U. A., Perveen, K., Hanif, F., and Simjee, S. U. (2014). N-(2-

hydroxy phenyl) acetamide produces profound inhibition of c-Fos protein and mRNA

expression in the brain of adjuvant-induced arthritic rats. Molecular and Cellular

Biochemistry, 387, 81-90.

Jawed, H., Shah, S. U. A., Jamall, S., and Simjee, S. U. (2010). N-(2-hydroxy phenyl)

acetamide inhibits inflammation-related cytokines and ROS in adjuvant-induced arthritic

(AIA) rats. International Immunopharmacology, 10, 900-905.

Jia, D., Koonce, N. A., Griffin, R. J., Jackson, C., and Corry, P. M. (2010). Prevention and

mitigation of acute death of mice after abdominal irradiation by the antioxidant N-acetyl-

cysteine (NAC). Radiation Research, 173, 579-589.

Jiang, C., Li, Z., Quan, H., Xiao, L., Zhao, J., Wang, Y., Liu, J., Gou, Y., An, S., and Huang,

Y. (2015). Osteoimmunology in orthodontic tooth movement. Oral Diseases, 21, 694-704.

Joosten, L. A., Helsen, M. M., Saxne, T., van de Loo, F. A., Heinegård, D., and van den Berg,

W. B. (1999). IL-1αβ blockade prevents cartilage and bone destruction in murine type II

Page 131: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

112

collagen-induced arthritis, whereas TNF-α blockade only ameliorates joint inflammation. The

Journal of Immunology, 163, 5049-5055.

Josien, R., Li, H. L., Ingulli, E., Sarma, S., Wong, B. R., Vologodskaia, M., Steinman, R. M.,

and Choi, Y. (2000). TRANCE, a tumor necrosis factor family member, enhances the

longevity and adjuvant properties of dendritic cells in vivo. Journal of Experimental Medicine,

191, 495-502.

Josien, R., Wong, B. R., Li, H.-L., Steinman, R. M., and Choi, Y. (1999). TRANCE, a TNF

family member, is differentially expressed on T cell subsets and induces cytokine production

in dendritic cells. The Journal of Immunology, 162, 2562-2568.

Kamalakkannan, N., and Prince, P. S. M. (2006). Rutin improves the antioxidant status in

streptozotocin-induced diabetic rat tissues. Molecular and Cellular Biochemistry, 293, 211-

219.

Kamphuis, S., Kuis, W., de Jager, W., Teklenburg, G., Massa, M., Gordon, G., Boerhof, M.,

Rijkers, G. T., Uiterwaal, C. S., and Otten, H. G. (2005). Tolerogenic immune responses to

novel T-cell epitopes from heat-shock protein 60 in juvenile idiopathic arthritis. The Lancet,

366, 50-56.

Karmakar, S., Kay, J., and Gravallese, E. M. (2010). Bone damage in rheumatoid arthritis:

mechanistic insights and approaches to prevention. Rheumatic Disease Clinics of North

America, 36, 385-404.

Kauss, T., Moynet, D., Rambert, J., Al-Kharrat, A., Brajot, S., Thiolat, D., Ennemany, R.,

Fawaz, F., and Mossalayi, M. D. (2008). Rutoside decreases human macrophage-derived

inflammatory mediators and improves clinical signs in adjuvant-induced arthritis. Arthritis

Research & Therapy, 10, p.R19.

Kawai, V. K., Stein, C. M., Perrien, D. S., and Griffin, M. R. (2012). Effects of anti-tumor

necrosis factor alpha agents on bone. Current Opinion in Rheumatology, 24, 576-585.

doi:10.1097/BOR.0b013e328356d212

Khan, M. M., Ahmad, A., Ishrat, T., Khuwaja, G., Srivastawa, P., Khan, M. B., Raza, S. S.,

Javed, H., Vaibhav, K., and Khan, A. (2009). Rutin protects the neural damage induced by

transient focal ischemia in rats. Brain Research, 1292, 123-135.

King, C. G., Kobayashi, T., Cejas, P. J., Kim, T., Yoon, K., Kim, G. K., Chiffoleau, E.,

Hickman, S. P., Walsh, P. T., and Turka, L. A. (2006). TRAF6 is a T cell–intrinsic negative

regulator required for the maintenance of immune homeostasis. Nature Medicine, 12, 1088-

1092.

Kirkham, B. W., Lassere, M. N., Edmonds, J. P., Juhasz, K. M., Bird, P. A., Lee, C. S., Shnier,

R., and Portek, I. J. (2006). Synovial membrane cytokine expression is predictive of joint

damage progression in rheumatoid arthritis: A two‐ year prospective study (the DAMAGE

study cohort). Arthritis & Rheumatism, 54, 1122-1131.

Page 132: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

113

Kitaura, H., Zhou, P., Kim, H.-J., Novack, D. V., Ross, F. P., and Teitelbaum, S. L. (2005).

M-CSF mediates TNF-induced inflammatory osteolysis. The Journal of Clinical

Investigation, 115, 3418-3427.

Klareskog, L., Rönnelid, J., Lundberg, K., Padyukov, L., and Alfredsson, L. (2008). Immunity

to citrullinated proteins in rheumatoid arthritis. Annual Review of Immunology., 26, 651-675.

Köhler, G., and Milstein, C. (1975). Continuous cultures of fused cells secreting antibody of

predefined specificity. Nature, 256, 495-497.

Komatsu, N., and Takayanagi, H. (2015). Arthritogenic T cells in autoimmune arthritis. The

International Journal of Biochemistry & Cell Biology, 58, 92-96.

Komine, M., Kukita, A., Kukita, T., Ogata, Y., Hotokebuchi, T., and Kohashi, O. (2001).

Tumor necrosis factor-α cooperates with receptor activator of nuclear factor κB ligand in

generation of osteoclasts in stromal cell-depleted rat bone marrow cell culture. Bone, 28, 474-

483.

Kong, Y. Y., Feige, U., Sarosi, I., Bolon, B., Tafuri, A., Morony, S., Capparelli, C., Li, J.,

Elliott, R., and McCabe, S. (1999). Activated T cells regulate bone loss and joint destruction

in adjuvant arthritis through osteoprotegerin ligand. Nature, 402, 43-47.

Kong, Y. Y., Yoshida, H., Sarosi, I., Tan, H. L., Timms, E., Capparelli, C., Morony, S.,

Oliveira-dos-Santos, A. J., Van, G., and Itie, A. (1999). OPGL is a key regulator of

osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature, 397,

315-323.

Kotake, S., Udagawa, N., Hakoda, M., Mogi, M., Yano, K., Tsuda, E., Takahashi, K., Furuya,

T., Ishiyama, S., and Kim, K. J. (2001). Activated human T cells directly induce

osteoclastogenesis from human monocytes: possible role of T cells in bone destruction in

rheumatoid arthritis patients. Arthritis & Rheumatism, 44, 1003-1012.

Kremer, J. M., Genovese, M. C., Cannon, G. W., Caldwell, J. R., Cush, J. J., Furst, D. E.,

Luggen, M. E., Keystone, E., Weisman, M. H., and Bensen, W. M. (2002). Concomitant

leflunomide therapy in patients with active rheumatoid arthritis despite stable doses of

methotrexate: a randomized, double-blind, placebo-controlled trial. Annals of Internal

Medicine, 137, 726-733.

Kudo, O., Fujikawa, Y., Itonaga, I., Sabokbar, A., Torisu, T., and Athanasou, N. A. (2002).

Proinflammatory cytokine (TNFα/IL‐ 1α) induction of human osteoclast formation. The

Journal of Pathology, 198, 220-227.

Kyung, T. W., Lee, J. E., Shin, H. H., and Choi, H. S. (2008). Rutin inhibits osteoclast

formation by decreasing reactive oxygen species and TNF-α by inhibiting activation of NF-

κB. Experimental & Molecular Medicine, 40, 52-58.

Page 133: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

114

La Casa, C., Villegas, I., De La Lastra, C. A., Motilva, V., and Calero, M. M. (2000). Evidence

for protective and antioxidant properties of rutin, a natural flavone, against ethanol induced

gastric lesions. Journal of Ethnopharmacology, 71, 45-53.

Labunskyy, V. M., and Gladyshev, V. N. (2013). Role of reactive oxygen species-mediated

signaling in aging. Antioxidants & Redox Signaling, 19, 1362-1372.

Lacey, D., Timms, E., Tan, H.-L., Kelley, M., Dunstan, C., Burgess, T., Elliott, R.,

Colombero, A., Elliott, G., and Scully, S. (1998). Osteoprotegerin ligand is a cytokine that

regulates osteoclast differentiation and activation. Cell, 93, 165-176.

Laferrère, B., García-Lorda, P., Russell, C. D., and Pi-Sunyer, F. X. (2004). Effect of oral

glucosamine sulfate on serum leptin levels in human subjects. Nutrition, 20, 321.

Lai, Y., Li, D., Li, C., Muehleisen, B., Radek, K. A., Park, H. J., Jiang, Z., Li, Z., Lei, H., and

Quan, Y. (2012). The antimicrobial protein REG3A regulates keratinocyte proliferation and

differentiation after skin injury. Immunity, 37, 74-84.

Lam, J., Takeshita, S., Barker, J. E., Kanagawa, O., Ross, F. P., and Teitelbaum, S. L. (2000).

TNF-α induces osteoclastogenesis by direct stimulation of macrophages exposed to

permissive levels of RANK ligand. The Journal of clinical investigation, 106, 1481-1488.

Lee, H., Lee, M. Y., Bhang, S. H., Kim, B. S., Kim, Y. S., Ju, J. H., Kim, K. S., and Hahn, S.

K. (2014). Hyaluronate–gold nanoparticle/tocilizumab complex for the treatment of

rheumatoid arthritis. ACS nano, 8, 4790-4798.

Lee, J. H., Jin, H., Shim, H. E., Kim, H. N., Ha, H., and Lee, Z. H. (2010). Epigallocatechin-

3-gallate inhibits osteoclastogenesis by down-regulating c-Fos expression and suppressing the

nuclear factor-κB signal. Molecular pharmacology, 77, 17-25.

Lee, Y. M., Fujikado, N., Manaka, H., Yasuda, H., and Iwakura, Y. (2010). IL-1 plays an

important role in the bone metabolism under physiological conditions. International

Immunology, 22, 805-816.

Li, P., Schwarz, E. M., O'Keefe, R. J., Ma, L., Looney, R. J., Ritchlin, C. T., Boyce, B. F., and

Xing, L. (2004). Systemic tumor necrosis factor α mediates an increase in peripheral

CD11bhigh osteoclast precursors in tumor necrosis factor α–transgenic mice. Arthritis &

Rheumatism, 50, 265-276.

Liu, Y., Li, M., He, Q., Yang, X., Ruan, F., and Sun, G. (2016). Periploca forrestii Saponin

Ameliorates Murine CFA-Induced Arthritis by Suppressing Cytokine Production. Mediators

of Inflammation, 2016.

Los, M., Dröge, W., Stricker, K., Baeuerle, P. A., and Schulze‐ Osthoff, K. (1995). Hydrogen

peroxide as a potent activator of T lymphocyte functions. European Journal of Immunology,

25, 159-165.

Page 134: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

115

Lubberts, E., van den Bersselaar, L., Oppers-Walgreen, B., Schwarzenberger, P., Coenen-de

Roo, C. J., Kolls, J. K., Joosten, L. A., and van den Berg, W. B. (2003). IL-17 promotes bone

erosion in murine collagen-induced arthritis through loss of the receptor activator of NF-κB

ligand/osteoprotegerin balance. The Journal of Immunology, 170, 2655-2662.

Ma, B., Dela Cruz, C. S., Hartl, D., Kang, M.-J., Takyar, S., Homer, R. J., Lee, C. G., and

Elias, J. A. (2011). RIG-like Helicase Innate Immunity Inhibits Vascular Endothelial Growth

Factor Tissue Responses via a Type I IFN–dependent Mechanism. American Journal of

Respiratory and Critical Care Medicine, 183, 1322-1335.

MacGregor, A. J., Snieder, H., Rigby, A. S., Koskenvuo, M., Kaprio, J., Aho, K., and Silman,

A. J. (2000). Characterizing the quantitative genetic contribution to rheumatoid arthritis using

data from twins. Arthritis & Rheumatism, 43, 30.

Mallick, I. H., Yang, W., Winslet, M. C., and Seifalian, A. M. (2004). REVIEW: Ischemia—

Reperfusion Injury of the Intestine and Protective Strategies Against Injury. Digestive

Diseases and Sciences, 49, 1359-1377.

Maruotti, N., Cantatore, F., Crivellato, E., Vacca, A., and Ribatti, D. (2007). Macrophages in

rheumatoid arthritis. Histology and histopathology: Cellular and Molecular Biology, 22, 581-

586.

McCartney-Francis, N., Allen, J. B., Mizel, D. E., Albina, J. E., Xie, Q., Nathan, C. F., and

Wahl, S. M. (1993). Suppression of arthritis by an inhibitor of nitric oxide synthase. Journal

of Experimental Medicine, 178, 749-754.

McCarty, D. J., and Carrera, G. F. (1982). Intractable rheumatoid arthritis: treatment with

combined cyclophosphamide, azathioprine, and hydroxychloroquine. The Journal of the

American Medical Association, 248, 1718-1723.

McCord, J. M., and Edeas, M. A. (2005). SOD, oxidative stress and human pathologies: a

brief history and a future vision. Biomedicine & Pharmacotherapy, 59, 139-142.

McHugh, J. M., and McHugh, W. B. (2000). Pain: neuroanatomy, chemical mediators, and

clinical implications. AACN Advanced Critical Care, 11, 168-178.

McInnes, I. B., and Schett, G. (2011). The pathogenesis of rheumatoid arthritis. New England

Journal of Medicine, 365, 2205-2219.

McNamee, K., Williams, R., and Seed, M. (2015). Animal models of rheumatoid arthritis:

How informative are they? European Journal of Pharmacology, 759, 278-286.

McNeill, E., Crabtree, M. J., Sahgal, N., Patel, J., Chuaiphichai, S., Iqbal, A. J., Hale, A. B.,

Greaves, D. R., and Channon, K. M. (2015). Regulation of iNOS function and cellular redox

state by macrophage Gch1 reveals specific requirements for tetrahydrobiopterin in NRF2

activation. Free Radical Biology and Medicine, 79, 206-216.

Page 135: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

116

Meyer, L.-H., Franssen, L., and Pap, T. (2006). The role of mesenchymal cells in the

pathophysiology of inflammatory arthritis. Best Practice & Research Clinical Rheumatology,

20, 969-981.

Miagkov, A. V., Kovalenko, D. V., Brown, C. E., Didsbury, J. R., Cogswell, J. P., Stimpson,

S. A., Baldwin, A. S., and Makarov, S. S. (1998). NF-κB activation provides the potential link

between inflammation and hyperplasia in the arthritic joint. Proceedings of the National

Academy of Sciences, 95, 13859-13864.

Mohandas, J., Duggin, G. G., Horvath, J. S., and Tiller, D. J. (1981). Metabolic oxidation of

acetaminophen (paracetamol) mediated by cytochrome P-450 mixed-function oxidase and

prostaglandin endoperoxide synthetase in rabbit kidney. Toxicology and Applied

Pharmacology, 61, 252-259.

Monaco, C., Nanchahal, J., Taylor, P., and Feldmann, M. (2015). Anti-TNF therapy: past,

present and future. International Immunology, 27, 55-62.

Moncada, S., and Higgs, A. (1993). The L-arginine-nitric oxide pathway. New England

Journal of Medicine, 329, 2002-2012.

Moreland, L. W., Pratt, P. W., Mayes, M. D., Postlethwaite, A., Weisman, M. H., Schnitzer,

T., Lightfoot, R., Calabrese, L., Zelinger, D. J., and Woody, J. N. (1995). Double‐ blind,

placebo‐ controlled multicenter trial using chimeric monoclonal anti‐ cd4 antibody, cm‐t412, in rheumatoid arthritis patients receiving concomitant methotrexate. Arthritis &

Rheumatology, 38, 1581-1588.

Mortaz, E., Lazar, Z., Koenderman, L., Kraneveld, A. D., Nijkamp, F. P., and Folkerts, G.

(2009). Cigarette smoke attenuates the production of cytokines by human plasmacytoid

dendritic cells and enhances the release of IL-8 in response to TLR-9 stimulation. Respiratory

Research, 10, 47.

Myers, D., Collier, F. M., Minkin, C., Wang, H., Holloway, W., Malakellis, M., and

Nicholson, G. (1999). Expression of functional RANK on mature rat and human osteoclasts.

FEBS letters, 463, 295-300.

Nabozny, G. H., Bull, M. J., Hanson, J., Griffiths, M. M., Luthra, H. S., and David, C. S.

(1994). Collagen-induced arthritis in T cell receptor V beta congenic B10. Q mice. The

Journal of Experimental Medicine, 180, 517-524.

Nagy, G., Clark, J. M., Buzás, E. I., Gorman, C. L., and Cope, A. P. (2007). Nitric oxide,

chronic inflammation and autoimmunity. Immunology Letters, 111, 1-5.

Nagy, G., Koncz, A., Telarico, T., Fernandez, D., Érsek, B., Buzás, E., and Perl, A. (2010).

Central role of nitric oxide in the pathogenesis of rheumatoid arthritis and sysemic lupus

erythematosus. Arthritis Research & Therapy, 12, 210. doi: 10.1186/ar3045.

Nakamura, H., Shibakawa, A., Tanaka, M., Kato, T., and Nishioka, K. (2003). Effects of

glucosamine hydrochloride on the production of prostaglandin E2, nitric oxide and

Page 136: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

117

metalloproteases by chondrocytes and synoviocytes in osteoarthritis. Clinical and

Experimental Rheumatology, 22, 293-299.

Nakashima, T., and Penninger, J. M. (2003). RANKL and RANK as novel therapeutic targets

for arthritis. Current Opinion in Rheumatology, 15, 280-287.

Nathan, C. (1992). Nitric oxide as a secretory product of mammalian cells. The FASEB

Journal, 6, 3051-3064.

Neumann, E., Gay, S., and Müller‐ Ladner, U. (2005). The RANK/RANKL/osteoprotegerin

system in rheumatoid arthritis: new insights from animal models. Arthritis & Rheumatology,

52, 2960-2967.

Nivsarkar, M. (2000). Improvement in circulating superoxide dismutase levels: role of

nonsteroidal anti-inflammatory drugs in rheumatoid arthritis. Biochemical and Biophysical

Research Communications, 270, 714-716.

O'dell, J. R., Leff, R., Paulsen, G., Haire, C., Mallek, J., Eckhoff, P. J., Fernandez, A., Blakely,

K., Wees, S., and Stoner, J. (2002). Treatment of rheumatoid arthritis with methotrexate and

hydroxychloroquine, methotrexate and sulfasalazine, or a combination of the three

medications: results of a two‐ year, randomized, double‐ blind, placebo‐ controlled trial.

Arthritis & Rheumatism, 46, 1164-1170.

Okamoto, H., Yoshio, T., Kaneko, H., and Yamanaka, H. (2010). Inhibition of NF‐ κB

signaling by fasudil as a potential therapeutic strategy for rheumatoid arthritis. Arthritis &

Rheumatism, 62, 82-92.

Onur, Ö., Akıncı, A., Akbıyık, F., and Ünsal, I. (2001). Elevated levels of nitrate in rheumatoid

arthritis. Rheumatology International, 20, 154-158.

Ostrakhovitch, E. A., and Afanas’ev, I. B. (2001). Oxidative stress in rheumatoid arthritis

leukocytes: suppression by rutin and other antioxidants and chelators. Biochemical

Pharmacology, 62, 743-746.

Palanki, M. (2002). Inhibitors of AP-1 and NF-κB mediated transcriptional activation:

therapeutic potential in autoimmune diseases and structural diversity. Current Medicinal

Chemistry, 9, 219-227.

Palma Zochio Tozzato, G., Taipeiro, E., Spadella, M., Marabini Filho, P., de Assis, M., Carlos,

C., Girol, A., and Chies, A. (2015). Collagen‐ induced arthritis increases inducible nitric

oxide synthase not only in aorta but also in the cardiac and renal microcirculation of mice.

Clinical & Experimental Immunology.

Palmer, R. M., Ferrige, A., and Moncada, S. (1987). Nitric oxide release accounts for the

biological activity of endothelium-derived relaxing factor. Nature, 327, 524-526.

Page 137: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

118

Panasyuk, A., Frati, E., Ribault, D., and Mitrovic, D. (1994). Effect of reactive oxygen species

on the biosynthesis and structure of newly synthesized proteoglycans. Free Radical Biology

and Medicine, 16, 157-167.

Pastore, A., Federici, G., Bertini, E., and Piemonte, F. (2003). Analysis of glutathione:

implication in redox and detoxification. Clinica Chimica Acta, 333, 19-39.

Patten, C. J., Thomas, P. E., Guy, R. L., Lee, M., Gonzalez, F. J., Guengerich, F. P., and Yang,

C. S. (1993). Cytochrome P450 enzymes involved in acetaminophen activation by rat and

human liver microsomes and their kinetics. Chemical Research in Toxicology, 6, 511-518.

Perveen, K., Hanif, F., Jawed, H., Jamall, S., and Simjee, S. U. (2014). N-(2-hydroxy phenyl)

acetamide: a novel suppressor of Toll-like receptors (TLR-2 and TLR-4) in adjuvant-induced

arthritic rats. Molecular and Cellular Biochemistry, 394, 67-75.

Pettit, A. R., Ji, H., Von Stechow, D., Müller, R., Goldring, S. R., Choi, Y., Benoist, C., and

Gravallese, E. M. (2001). TRANCE/RANKL knockout mice are protected from bone erosion

in a serum transfer model of arthritis. The American Journal of Pathology, 159, 1689-1699.

Pham, T. N., Rahman, P., Tobin, Y. M., Khraishi, M. M., Hamilton, S. F., Alderdice, C., and

Richardson, V. J. (2003). Elevated serum nitric oxide levels in patients with inflammatory

arthritis associated with co-expression of inducible nitric oxide synthase and protein kinase

C-eta in peripheral blood monocyte-derived macrophages. The Journal of Rheumatology, 30,

2529-2534.

Picerno, V., Ferro, F., Adinolfi, A., Valentini, E., Tani, C., and Alunno, A. (2015). One year

in review: the pathogenesis of rheumatoid arthritis. Clinical and Experimental Rheumatology,

33, 551-558.

Potter, W., Davis, D., Mitchell, J., Jollow, D., Gillette, J., and Brodie, B. (1973).

Acetaminophen-induced hepatic necrosis. III. Cytochrome P-450-mediated covalent binding

in vitro. Journal of Pharmacology and Experimental Therapeutics, 187, 203-210.

Radad, K., Al-Shraim, M., Moldzio, R., and Rausch, W. D. (2012). Recent advances in

benefits and hazards of engineered nanoparticles. Environmental Toxicology and

Pharmacology, 34, 661-672.

Rao D. S. (2015) Rheumatoid arthritis (RA) disease treatment with rutin stabilized

nanoparticles. Austin Journal of Biotechnology & Bioengineering, 2, 1043.

Redlich, K., Hayer, S., Maier, A., Dunstan, C. R., Tohidast‐ Akrad, M., Lang, S., Türk, B.,

Pietschmann, P., Woloszczuk, W., and Haralambous, S. (2002). Tumor necrosis factor α‐mediated joint destruction is inhibited by targeting osteoclasts with osteoprotegerin. Arthritis

& Rheumatism, 46, 785-792.

Regan, E., Bowler, R., and Crapo, J. (2008). Joint fluid antioxidants are decreased in

osteoarthritic joints compared to joints with macroscopically intact cartilage and subacute

injury. Osteoarthritis and Cartilage, 16, 515-521.

Page 138: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

119

Reinhart, K., Wiegand-Lohnert, C., Grimminger, F., Kaul, M., Withington, S., Treacher, D.,

Eckart, J., Willatts, S., Bouza, C., and Krausch, D. (1996). Assessment of the safety and

efficacy of the monoclonal anti-tumor necrosis factor antibody-fragment, MAK 195F, in

patients with sepsis and septic shock: a multicenter, randomized, placebo-controlled, dose-

ranging study. Critical care medicine, 24, 733-742.

Roessner, A., Kuester, D., Malfertheiner, P., and Schneider-Stock, R. (2008). Oxidative stress

in ulcerative colitis-associated carcinogenesis. Pathology-Research and Practice, 204, 511-

524.

Rojkind, M., Dominguez-Rosales, J.-A., Nieto, N., and Greenwel, P. (2002). Role of hydrogen

peroxide and oxidative stress in healing responses. Cellular and Molecular Life Sciences, 59,

1872-1891.

Romas, E., Sims, N. A., Hards, D. K., Lindsay, M., Quinn, J. W., Ryan, P. F., Dunstan, C. R.,

Martin, T. J., and Gillespie, M. T. (2002). Osteoprotegerin reduces osteoclast numbers and

prevents bone erosion in collagen-induced arthritis. The American Journal of Pathology, 161,

1419-1427.

Rosloniec, E. F., Cremer, M., Kang, A., and Myers, L. K. (2001). Collagen-induced arthritis.

Current Protocols in Immunology, Chapter 15, Unit 15.5.

doi:10.1002/0471142735.im1505s20.

Ruderman, E. M. (2012). Overview of safety of non-biologic and biologic DMARDs.

Rheumatology, 51, vi37-vi43. doi: https://doi.org/10.1093/rheumatology/kes283.

Ruocco, M. G., Maeda, S., Park, J. M., Lawrence, T., Hsu, L. C., Cao, Y., Schett, G., Wagner,

E. F., and Karin, M. (2005). IκB kinase (IKK)β, but not IKKα, is a critical mediator of

osteoclast survival and is required for inflammation-induced bone loss. Journal of Cell

Biology. 201, 1677–1687. doi: 10.1084/jem.20042081.

Saeed, S. A., Saeed, S. O. (1975). Patents (Inventions). US Patent No, 3, 758.

Saag, K. G., Teng, G. G., Patkar, N. M., Anuntiyo, J., Finney, C., Curtis, J. R., Paulus, H. E.,

Mudano, A., Pisu, M., and Elkins‐ Melton, M. (2008). American College of Rheumatology

2008 recommendations for the use of nonbiologic and biologic disease‐ modifying

antirheumatic drugs in rheumatoid arthritis. Arthritis Care & Research, 59, 762-784.

Santana-Sabagun, E., and Weisman, M. H. (2001). Nonsteroidal anti-inflammatory drugs. In:

Ruddy S, Harris JED, Sledge., C. B, Budd., R.C, Sergent JS., (Eds.). Kelly’s Textbook of

Rheumatology. 1, 799-822. Philadelphia, Pa, USA: W.B. Saunders Company.

Sanvicens, N., and Marco, M. P. (2008). Multifunctional nanoparticles–properties and

prospects for their use in human medicine. Trends in Biotechnology, 26, 425-433.

Scheinman, R. I., Trivedi, R., Vermillion, S., and Kompella, U. B. (2011). Functionalized

STAT1 siRNA nanoparticles regress rheumatoid arthritis in a mouse model. Nanomedicine,

6, 1669-1682.

Page 139: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

120

Scher, J. U., and Abramson, S. B. (2011). The microbiome and rheumatoid arthritis. Nature

Reviews Rheumatology, 7, 569-578.

Schett, G., Hayer, S., Zwerina, J., Redlich, K., and Smolen, J. S. (2005). Mechanisms of

disease: the link between RANKL and arthritic bone disease. Nature Clinical Practice

Rheumatology, 1, 47-54.

Scott, D. L., and Wolfe, F. (2010). Huizinga TW. Rheumatoid arthritis. Lancet, 376, 1094-

1108.

Scott, D., and Kingsley, G. (2006). Tumor necrosis factor inhibitors for rheumatoid arthritis.

New England Journal of Medicine, 355, 704-712.

Sen, C. K., and Roy, S. (2008). Redox signals in wound healing. Biochimica et Biophysica

Acta (BBA)-General Subjects, 1780, 1348-1361.

Shalhoub, V., Elliott, G., Chiu, L., Manoukian, R., Kelley, M., Hawkins, N., Davy, E.,

Shimamoto, G., Beck, J., and Kaufman, S. (2000). Characterization of osteoclast precursors

in human blood. British Journal of Haematology, 111, 501-512.

Shaulian, E., and Karin, M. (2002). AP-1 as a regulator of cell life and death. Nature Cell

Biology, 4, E131-E136. doi: 10.1038/ncb0502-e131.

Shiozawa, S., Shimizu, K., Tanaka, K., and Hino, K. (1997). Studies on the contribution of c-

fos/AP-1 to arthritic joint destruction. Journal of Clinical Investigation, 99, 1210.

Simonet, W., Lacey, D., Dunstan, C., Kelley, M., Chang, M.-S., Lüthy, R., Nguyen, H.,

Wooden, S., Bennett, L., and Boone, T. (1997). Osteoprotegerin: a novel secreted protein

involved in the regulation of bone density. Cell, 89, 309-319.

Simsa-Maziel, S., Zaretsky, J., Reich, A., Koren, Y., Shahar, R., and Monsonego-Ornan, E.

(2013). IL-1RI participates in normal growth plate development and bone modeling.

American Journal of Physiology-Endocrinology and Metabolism, 305, E15-E21.

Sköldstam, L., Hagfors, L., and Johansson, G. (2003). An experimental study of a

Mediterranean diet intervention for patients with rheumatoid arthritis. Annals of the

Rheumatic Diseases, 62, 208-214.

Smith, J. B., and Haynes, M. K. (2002). Rheumatoid arthritis—a molecular understanding.

Annals of Internal Medicine, 136, 908-922.

Soneja, A., Drews, M., and Malinski, T. (2005). Role of nitric oxide, nitroxidative and

oxidative stress in wound healing. Pharmacological Reports, 57, 108.

Stamp, L. K., Khalilova, I., Tarr, J. M., Senthilmohan, R., Turner, R., Haigh, R. C., Winyard,

P. G., and Kettle, A. J. (2012). Myeloperoxidase and oxidative stress in rheumatoid arthritis.

Rheumatology, 51, 1796-1803.

Page 140: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

121

Stenström, C. H., and Minor, M. A. (2003). Evidence for the benefit of aerobic and

strengthening exercise in rheumatoid arthritis. Arthritis Care & Research, 49, 428-434.

Stolt, P., Bengtsson, C., Nordmark, B., Lindblad, S., Lundberg, I., Klareskog, L., and

Alfredsson, L. (2003). Quantification of the influence of cigarette smoking on rheumatoid

arthritis: results from a population based case-control study, using incident cases. Annals of

the Rheumatic Diseases, 62, 835-841.

Svensson, B., Boonen, A., Albertsson, K., Van der Heijde, D., Keller, C., and Hafström, I.

(2005). Low‐ dose prednisolone in addition to the initial disease‐ modifying antirheumatic

drug in patients with early active rheumatoid arthritis reduces joint destruction and increases

the remission rate: A two‐ year randomized trial. Arthritis & Rheumatism, 52, 3360-3370.

Symons, J., McDowell, T., Di Giovine, F., Wood, N., Capper, S., and Duff, G. (1988).

Interleukin 1 in rheumatoid arthritis: potentiation of immune responses within the joint.

Lymphokine Research, 8, 365-372.

Tak, P. P., and Firestein, G. S. (2001). NF-κB: a key role in inflammatory diseases. The

Journal of Clinical Investigation, 107, 7-11.

Takahashi, N., Akatsu, T., Udagawa, N., Sasaki, T., Yamaguchi, A., Moseley, J. M., Martin,

T. J., and Suda, T. (1988). Osteoblastic cells are involved in osteoclast formation.

Endocrinology, 123, 2600-2602.

Takayanagi, H. (2005). Mechanistic insight into osteoclast differentiation in

osteoimmunology. Journal of Molecular Medicine, 83, 170-179.

Tanaka, S., Matsui, T., Murakami, T., Ishizuka, T., Sugiura, M., Kawashima, K., and Sugita,

T. (1998). Immunological abnormality associated with the augmented nitric oxide synthesis

in adjuvant-induced arthritis. International Journal of Immunopharmacology, 20, 71-84.

Tang, D., Shi, Y., Kang, R., Li, T., Xiao, W., Wang, H., and Xiao, X. (2007). Hydrogen

peroxide stimulates macrophages and monocytes to actively release HMGB1. Journal of

Leukocyte Biology, 81, 741-747.

Teitelbaum, S. L. (2004). RANKing c-Jun in osteoclast development. The Journal of Clinical

Investigation, 114, 463-465.

Tracey, D., Klareskog, L., Sasso, E. H., Salfeld, J. G., and Tak, P. P. (2008). Tumor necrosis

factor antagonist mechanisms of action: a comprehensive review. Pharmacology &

Therapeutics, 117, 244-279.

Trentham, D. E. (1982). Collagen arthritis as a relevant model for rheumatoid arthritis.

Evidence pro and con. Arthritis & Rheumatology, 25, 911-916.

Trentham, D. E., Townes, A. S., and Kang, A. H. (1977). Autoimmunity to type II collagen

an experimental model of arthritis. Journal of Experimental Medicine, 146, 857-868.

Page 141: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

122

Tsao, P. W., Suzuki, T., Totsuka, R., Murata, T., Takagi, T., Ohmachi, Y., Fujimura, H., and

Takata, I. (1997). The effect of dexamethasone on the expression of activated NF-κB in

adjuvant arthritis. Clinical Immunology and Immunopathology, 83, 173-178.

Tsubaki, M., Takeda, T., Kino, T., Itoh, T., Imano, M., Tanabe, G., Muraoka, O., Satou, T.,

and Nishida, S. (2015). Mangiferin suppresses CIA by suppressing the expression of TNF-α,

IL-6, IL-1β, and RANKL through inhibiting the activation of NF-κB and ERK1/2. American

Journal of Translational Research, 7, 1371.

Tugwell, P., Pincus, T., Yocum, D., Stein, M., Gluck, O., Kraag, G., McKendry, R., Tesser,

J., Baker, P., and Wells, G. (1995). Combination therapy with cyclosporine and methotrexate

in severe rheumatoid arthritis. New England Journal of Medicine, 333, 137-142.

Umar, S., Mishra, N. K., Pal, K., Sajad, M., Ansari, M. M., Ahmad, S., Katiyar, C. K., and

Khan, H. A. (2012). Protective effect of rutin in attenuation of collagen-induced arthritis in

Wistar rat by inhibiting inflammation and oxidative stress. Indian Journal of Rheumatology,

7, 191-198.

Valko, M., Leibfritz, D., Moncol, J., Cronin, M. T., Mazur, M., and Telser, J. (2007). Free

radicals and antioxidants in normal physiological functions and human disease. The

International Journal of Biochemistry & Cell Biology, 39, 44-84.

Van der Heijden, J., Oerlemans, R., Lems, W., Scheper, R., Dijkmans, B., and Jansen, G.

(2009). The proteasome inhibitor bortezomib inhibits the release of NFkappaB-inducible

cytokines and induces apoptosis of activated T cells from rheumatoid arthritis patients.

Clinical and Experimental Rheumatology, 27, 92-98.

Van der Leeden, M., Steultjens, M., Dekker, J., Prins, A., and Dekker, J. (2006). Forefoot

joint damage, pain and disability in rheumatoid arthritis patients with foot complaints: the role

of plantar pressure and gait characteristics. Rheumatology, 45, 465-469.

Van der Lubbe, P. A., Dijkmans, B. A., Markusse, H. M., Nässander, U., and Breedveld, F.

C. (1995). A randomized, double‐ blind, placebo‐ controlled study of CD4 monoclonal

antibody therapy in early rheumatoid arthritis. Arthritis & Rheumatism, 38, 1097-1106.

Van Tuyl, L. H., Voskuyl, A. E., Boers, M., Geusens, P., Landewé, R. B., Dijkmans, B. A.,

and Lems, W. F. (2010). Baseline RANKL: OPG ratio and markers of bone and cartilage

degradation predict annual radiological progression over 11 years in rheumatoid arthritis.

Annals of the Rheumatic Diseases, 69, 1623-8. doi: 10.1136/ard.2009.121764.

Van Vollenhoven, R. F. (2015). Biologics for the Treatment of Rheumatoid Arthritis. Springer. doi: 10.1007/978-3-319-13108-5_2.

Van'T Hof, R. J., and Ralston, S. H. (2001). Nitric oxide and bone. Immunology, 103, 255-

261.

Vasanthi, P., Nalini, G., and Rajasekhar, G. (2009). Status of oxidative stress in rheumatoid

arthritis. International Journal of Rheumatic Diseases, 12, 29-33.

Page 142: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

123

Vervoordeldonk, M. J., and Tak, P. P. (2002). Cytokines in rheumatoid arthritis. Current

Rheumatology Reports, 4, 208-217.

Vidal, N., Brandstrom, H., Jonsson, K., and Ohlsson, C. (1998). Osteoprotegerin mRNA is

expressed in primary human osteoblast-like cells: down-regulation by glucocorticoids.

Journal of Endocrinology, 159, 191-195.

Vincent, C., De Keyser, F., Masson-Bessière, C., Sebbag, M., Veys, E. M., and Serre, G.

(1999). Anti-perinuclear factor compared with the so called “antikeratin” antibodies and

antibodies to human epidermis filaggrin, in the diagnosis of arthritides. Annals of the

Rheumatic Diseases, 58, 42-48.

Vincent, T. L., Williams, R. O., Maciewicz, R., Silman, A., Garside, P., and Group, A. R. U.

A. M. W. (2012). Mapping pathogenesis of arthritis through small animal models.

Rheumatology, 51, 1931-41. doi: 10.1093/rheumatology/kes035.

Wagner, E. F., and Eferl, R. (2005). Fos/AP‐ 1 proteins in bone and the immune system.

Immunological Reviews, 208, 126-140.

Wakamatsu, K., Nanki, T., Miyasaka, N., Umezawa, K., and Kubota, T. (2005). Effect of a

small molecule inhibitor of nuclear factor-κB nuclear translocation in a murine model of

arthritis and cultured human synovial cells. Arthritis Research & Therapy, 7, R1348.

Wållberg-Jonsson, S., Johansson, H., Ohman, M., and Rantapää-Dahlqvist, S. (1999). Extent

of inflammation predicts cardiovascular disease and overall mortality in seropositive

rheumatoid arthritis. A retrospective cohort study from disease onset. The Journal of

Rheumatology, 26, 2562-2571.

Walters, M., and Cawley, M. (1988). Combined suppressive drug treatment in severe

refractory rheumatoid disease: an analysis of the relative effects of parenteral

methylprednisolone, cyclophosphamide, and sodium aurothiomalate. Annals of the Rheumatic

Diseases, 47, 924-929.

Wassenberg, S., Rau, R., Steinfeld, P., and Zeidler, H. (2005). Very low‐ dose prednisolone

in early rheumatoid arthritis retards radiographic progression over two years: a multicenter,

double‐ blind, placebo‐ controlled trial. Arthritis & Rheumatism, 52, 3371-3380.

Watson, D. J., Rhodes, T., and Guess, H. A. (2003). All-cause mortality and vascular events

among patients with rheumatoid arthritis, osteoarthritis, or no arthritis in the UK General

Practice Research Database. The Journal of Rheumatology, 30, 1196-1202.

Wei, S., Kitaura, H., Zhou, P., Ross, F. P., and Teitelbaum, S. L. (2005). IL-1 mediates TNF-

induced osteoclastogenesis. The Journal of Clinical Investigation, 115, 282-290.

Wei, Z. f., Tong, B., Xia, Y.-f., Lu, Q., Chou, G.-x., Wang, Z.-t., and Dai, Y. (2013).

Norisoboldine suppresses osteoclast differentiation through preventing the accumulation of

TRAF6-TAK1 complexes and activation of MAPKs/NF-κB/c-Fos/NFATc1 Pathways. PloS

one, 8, e59171. doi:10.1371/journal.pone.0059171.

Page 143: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

124

Weinblatt, M. E., Dixon, J. A., and Falchuk, K. R. (2000). Serious liver disease in a patient

receiving methotrexate and leflunomide. Arthritis & Rheumatology, 43, 2609-2611.

Weinblatt, M. E., Kremer, J. M., Coblyn, J. S., Maier, A. L., Helfgott, S. M., Morrell, M.,

Byrne, V. M., Kaymakcian, M. V., and Strand, V. (1999). Pharmacokinetics, safety, and

efficacy of combination treatment with methotrexate and leflunomide in patients with active

rheumatoid arthritis. Arthritis & Rheumatism, 42, 1322-1328.

Wieland, H. A., Michaelis, M., Kirschbaum, B. J., and Rudolphi, K. A. (2005).

Osteoarthritis—an untreatable disease? Nature reviews Drug discovery, 4, 331-344.

Wiesenthal, A., Hunter, L., Wang, S., Wickliffe, J., and Wilkerson, M. (2011). Nanoparticles:

small and mighty. International Journal of Dermatology, 50, 247-254.

Willkens, R. F., Urowitz, M. B., Stablein, D. M., Mckendry, R. J., Berger, R. G., Box, J. H.,

Fiechtner, J. J., Fudman, E. J., Paul Hudson, N., and Marks, C. R. (1992). Comparison of

azathioprine, methotrexate, and the combination of both in the treatment of rheumatoid

arthritis. Arthritis & Rheumatism, 35, 849-856.

Wilske, K., and Healey, L. (1989). Remodeling the pyramid--a concept whose time has come.

The Journal of Rheumatology, 16, 565-567.

Winyard, P. G., Ryan, B., Eggleton, P., Nissim, A., Taylor, E., Faro, M. L. L., Burkholz, T.,

Szabó-Taylor, K. E., Fox, B., and Viner, N. Haigh, R. C., Benjamin, N., Jones, A. M.,

Whiteman, M. (2011). Measurement and meaning of markers of reactive species of oxygen,

nitrogen and sulfur in healthy human subjects and patients with inflammatory joint disease.

Biochemical Society Transactions, 39, 1226-32. doi: 10.1042/BST0391226.

Wollenhaupt, J., and Zeidler, H. (1997). Combined administration of long-acting

antirheumatic drugs in the treatment of chronic polyarthritis. Dtsch Med Wochenschr, 122,

1219-1223. doi:10.1055/s-2008-1047751

Wong, B. R., Besser, D., Kim, N., Arron, J. R., Vologodskaia, M., Hanafusa, H., and Choi,

Y. (1999). TRANCE, a TNF family member, activates Akt/PKB through a signaling complex

involving TRAF6 and c-Src. Molecular cell, 4, 1041-1049.

Wong, B. R., Rho, J., Arron, J., Robinson, E., Orlinick, J., Chao, M., Kalachikov, S., Cayani,

E., Bartlett, F. S., and Frankel, W. N. (1997). TRANCE is a novel ligand of the tumor necrosis

factor receptor family that activates c-Jun N-terminal kinase in T cells. Journal of Biological

chemistry, 272, 25190-25194.

Wong, S. H., and Lord, J. M. (2004). Factors underlying chronic inflammation in rheumatoid

arthritis. Archivum Immunologiae et Therapiae Experimentalis, 52, 379-388.

Wu, D., Cederbaum, A. I. (2009). Oxidative stress and alcoholic liver disease. Seminars in

Liver Disease, 29, 141–54.

Yamazaki, K., Fukuda, K., Matsukawa, M., Hara, F., Matsushita, T., Yamamoto, N., Yoshida,

K., Munakata, H., and Hamanishi, C. (2003). Cyclic tensile stretch loaded on bovine

Page 144: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

125

chondrocytes causes depolymerization of hyaluronan: involvement of reactive oxygen

species. Arthritis & Rheumatism, 48, 3151-3158.

Yamazaki, K., Fukuda, K., Matsukawa, M., Hara, F., Yoshida, K., Akagi, M., Munakata, H.,

and Hamanishi, C. (2003). Reactive oxygen species depolymerize hyaluronan: involvement

of the hydroxyl radical. Pathophysiology, 9, 215-220.

Yu, D. H., Yi, J. K., Yuh, H. S., jin Park, S., Kim, H. J., Bae, K. B., Ji, Y. R., Kim, N. R.,

Park, S. J., and Kim, D. H. (2012). Over-expression of extracellular superoxide dismutase in

mouse synovial tissue attenuates the inflammatory arthritis. Experimental & Molecular

Medicine, 44, 529-535.

Zhang, L., Yu, M., Deng, J., Lv, X., Liu, J., Xiao, Y., Yang, W., Zhang, Y., and Li, C. (2015).

Chemokine Signaling Pathway Involved in CCL2 Expression in Patients with Rheumatoid

Arthritis. Yonsei Medical Journal, 56, 1134-1142.

Zhou, H. f., Yan, H., Hu, Y., Springer, L. E., Yang, X., Wickline, S. A., Pan, D., Lanza, G.

M., and Pham, C. T. (2014). Fumagillin prodrug nanotherapy suppresses macrophage

inflammatory response via endothelial nitric oxide. ACS nano, 8, 7305-7317.

Zolnik, B. S., Gonzalez-Fernandez, A., Sadrieh, N., and Dobrovolskaia, M. A. (2010).

Minireview: nanoparticles and the immune system. Endocrinology, 151, 458-465.

Page 145: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

126

PERSONAL INTRODUCTION

Anum Gul

E.mail: [email protected]

I was born in Karachi and completed my secondary school certificate from Khatoon-e-

Pakistan Govt. Girls Secondary School, Karachi in 2004. After matriculation, I completed my

higher secondary school certificate from Khatoon-e-Pakistan Govt. Degree College for

Women, Karachi in 2006. I completed B.Sc. (Hons) in Biotechnology in First class, First

Position from University of Karachi in 2009. In 2010, I received Gold medal for getting First

class “First Position” in my Master’s degree in Biotechnology from University of Karachi.

Since my childhood, whenever, I was asked about my future profession, I always said that I

want to be a scientist and during my masters, I was sure that I will do higher studies and will

do my best in the field of science. To continue my dream, I joined one of the best research

institute, Dr. Panjwani Center for Molecular Medicine and Drug Research, International

Center for Chemical and Biological Sciences, University of Karachi for getting Ph.D. in

molecular medicine in 2011.

Initially, I worked under the supervision of Dr. Talat Makhmoor till 2015 then I joined Dr.

Shabana U. Simjee and I was lucky to get training from both the supervisors. During my entire

stay in PCMD, I learned different molecular and cell biology techniques, attended and

participated as a volunteer in various seminars and international symposia held at my institute,

where I got experience to meet the world-renowned scientists. I also got opportunity to teach

microbiology in GRE preparatory classes held in PCMD. I thoroughly enjoyed every

academic and research activity during my Ph.D. studies.

Page 146: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

127

GLOSSARY

Adjuvant: Any substance which enhance the efficacy of vaccine most commonly by

increasing their half-life.

Angiogenesis: the formation of new blood vessels, especially those that supply oxygen and

nutrients to cancerous tissues.

Anti-citrullinated protein antibodies: are autoantibodies (antibodies directed against one or

more of an individual’s own proteins) that are frequently detected in the blood of rheumatoid

arthritis patients. The main epitope for these antibodies is filaggrin.

Antigen: Any particular substance that provokes immune system to produce specific

antibodies against it.

B-cells: a lymphocyte derived from bone marrow that provides humoral immunity, it

recognizes free antigen molecules and matures into plasma cells that secrete antibodies, which

inactivate the antigens.

c-Fos: DNA-binding proteins encoded by the c-fos genes, involved in growth related

transcriptional control. It combines with c-jun to form c-fos/c-jun heterodimer (transcription

factor AP-1).

Chemokines: a large family of cytokines having chemokinetic and chemotactic properties,

known to stimulate leukocyte movement and attraction.

Chondrocytes: a connective tissue cell that occupies a lacuna (small cavities) within the

cartilage matrix also called cartilage cell.

Page 147: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

128

Complement: a substance that is produced by a precursor protein or in response to the

presence of foreign material in the body that triggers or participates in a complement reaction.

c-Jun: is a protein encoded by proto oncogene c-jun that helps in cell differentiation, cell

growth and apoptosis.

Immunization: It’s an immunological procedure in which an organism’s immune system is

provoked against a particular antigen using immunogen.

Infection: It’s a condition that potentiates abnormal immune responses due to invasion of

microorganisms, resulting in serious pathological problems.

Leukotrienes: a class of small molecules produced by cells in response to allergen exposure,

they are the regulators of inflammatory and allergic reactions.

Mitogen: is a chemical substance usually protein that encourages a cell to commence cell

division, triggering mitosis it triggers signal transduction pathways in which mitogen activated

protein kinase is involved, leading to mitosis.

MMPs: enzymes related to tissue healing/remodeling and cancer cell metastasis.

Osteoblast: a cell from which bone develops or a bone-forming cell.

Osteoclast: a cell that absorbs bone.

Plasmin: an enzyme belongs to serine proteases, released as a zymogen called plasminogen

and converted into active plasmin by various enzymes. It is present in blood that degrades

many blood plasma proteins including fibrin, fibronectin, thrombospondin and laminin etc.

also known to activate collagenases, some mediators of complement system.

Page 148: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

129

Prostaglandlins: any of a group of naturally occurring, chemically related fatty acids that

stimulate contractility of the uterine and other smooth muscle and have the ability to lower

blood pressure, regulate acid secretion of the stomach, body temperature, platelet aggregation,

and control inflammation and vascular permeability; they also affect the action of certain

hormones.

Proteoglycans: are proteins that are heavily glycosylated. The basic proteoglycan unit

consists of a "core protein" with one or more covalently attached glycosaminoglycan chains.

The chains are long, linear carbohydrate polymers that are negatively charged under

physiological conditions, due to the occurrence of sulfate and uronic acid groups.

Proteoglycans occur in the connective tissue.

T-cells: a small lymphocyte developed in the thymus; it orchestrates the immune system's

response to infected or malignant cells.

Page 149: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

130

APPENDIX I

S.No. Name of Chemical Source Catalogue #

1. Chloroform Fischer Scientific C/4960/17

2. DAPI Wako 049-18801

3. DEPC Carl Roth K028.3

4. Dibasic Sodium Phosphate, Na2HPO4.2H2O Merck 1.065576

5. DMSO Wako 042-21765

6. DPX mounting medium Scharlau DP0050

7. Ethanol Fisher 64-17-5

8. Formalin Scharlau FO 0010

9. Isopropanol Tedia PR-1490

10. Monobasic Sodium Phosphate, NaH2PO4.2H2O Merck 1.06345

11. Paraffin Carl Roth Paraplast Plus

12. PEG-400 Scharlau PO0035

13. Roti-Histofix Carl Roth P087.1

14. Trizol Life Technologies 15596-026

15. Xylene Scharlau X10057

Page 150: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

131

List of Staining Reagents

S.No. Name of Chemical Source Catalogue No.

1. 0.5% Eosin Y solution (Eosin G) Carl Roth X883.1

2. Hematoxylin Carl Roth T864.2

List of Instruments

S.No. Instrument Company

1. Centrifuge 5810R Eppendorf

2. Chemical Fume hood Lab Tech

3. Electronic Balance Sartorius, CP225D

4. Homogenizer Ultra-Turrax, T8, IKA, Germany

5. Hot plate drier for slides Medite

6. Incubator LabTech

7. Magnetic Hotplate stirrer Lab Tech

8. Master Cycler Pro S PCR Machine Eppendorf

9. Microtome Yidi

10. Nanodrop 2000

Spectrophotometer

ThermoScientific

11. Oven LabTech

Page 151: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

132

12. Paraffin Embedding System Kedee

13. Periplastic Pump Eyela

14. pH meter Jenway

15. Real Time PCR Machine Agilent Technologies Stratagene Mx3000P

16. Surgical Instruments Local

17. Upright multichannel fluorescence

microscope

90i, Nikon Corporation, Japan

18. Vortex Mixer Lab Tech

19. Water bath LabTech

20. Water Purification System Millipore, Elix 5, Simplicity

Page 152: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

133

APPENDIX II

1. Phosphate Buffer Saline

(a) 0.1 M Mono Basic Phosphate NaH2PO4.2H2O (Mol. Wt 156)

Dissolve 15.6gm in 1 L of 18.2 MΩ water to make 0.1M solution. Store this stock solution

at room temperature.

(b) 0.1M Di Basic Phosphate Na2HPO4.2H2O (Mol Wt. 178)

Dissolve 17.8gm in 1 L of 18.2 MΩ water to make 0.1 M solution. Put 1 L water in a beaker

and start stirring. Then add a small amount of Dibasic phosphate. Make sure to dissolve the

whole amount and only then add some more Dibasic phosphate and dissolve it. In this manner

add Dibasic phosphate gradually until the whole amount is dissolved. Store this stock solution

at room temperature. Never put the stock solution in the freeze as the Dibasic phosphate will

crash out of the solution.

(c) 0.1 M PB preparation

Take 500 mL 0.1 M Dibasic phosphate in a beaker. Put the beaker on the hotplate magnetic

stirrer. Put the electrode of the pH meter and the temperature sensitive probe in the solution

and start stirring at a medium speed. Add 0.1 M Mono basic phosphate until pH reaches 7.3.

Usually ~280 mL Mono is needed for 500 mL Di.

(d) 0.05 M PBS

Take 500 ml of 0.1 M PB and 500 ml of water 18.2 MΩ in 1L graduated cylinder. Add 9gm

of NaCl and stir through magnetic stirrer. Transfer in a clean and labeled 1L bottle and keep

at room temperature.

2. Formaline Fixative (3.7%) or Neutral Buffered Formaline (NBF)

1. Dissolve 200ml of Phosphate buffer (0.1 M), 40ml Formaline (37%) and 160ml of

simplicity water 18.2M in a beaker through magnetic stirrer.

2. Keep it at room temperature.

3. Decalcification Medium

Formalin (37%) 90 mL

Formic acid 10 mL

Page 153: Suppression of Collagen Induced Arthritis by NA-2 and ...prr.hec.gov.pk/jspui/bitstream/123456789/8042/1/... · CERTIFICATE It is certified that the thesis entitled, “Suppression

134

LIST OF PUBLICATIONS

1. Gul, A., Kunwar, B., Mazhar, M., Perveen, K., Simjee, S. U. (2017). N-(2-

Hydroxyphenyl) acetamide: a Novel Suppressor of RANK/RANKL Pathway in

Collagen-Induced Arthritis Model in Rats. Inflammation. doi: 10.1007/s10753-017-

0561-1.

2. Mazhar, M., Faizi, S., Gul, A., Kabir, N., Simjee, S. U. (2017). Effects of naturally

occurring flavonoids on ferroportin expression in the spleen in iron deficiency anemia

in vivo. RSC Advances. doi: 10.1039/c7ra02138k.