165
Functional characterisation of UPOX1, a mitochondrial stress responsive protein in Arabidopsis thaliana . Vindya Uggalla, BSc (Hons) This thesis is presented for the degree of Doctor of Philosophy of The University of Western Australia School of Chemistry and Biochemistry ARC Centre of Excellence for Plant Energy Biology 2017

Functional characterisation of UPOX1, a mitochondrial ... · ARC Centre of Excellence for Plant Energy Biology 2017 . 2 THESIS DECLARATION I, Vindya Uggalla, certify that: ... 2.7.3

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Functional characterisation of UPOX1, a

mitochondrial stress responsive protein in

Arabidopsis thaliana.

Vindya Uggalla, BSc (Hons)

This thesis is presented for the degree of Doctor of Philosophy of The University of Western Australia

School of Chemistry and Biochemistry

ARC Centre of Excellence for Plant Energy Biology

2017

2

THESIS DECLARATION

I, Vindya Uggalla, certify that:

This thesis has been substantially accomplished during enrolment in the degree.

This thesis does not contain material which has been accepted for the award of any other degree or diploma in my name, in any university or other tertiary institution.

No part of this work will, in the future, be used in a submission in my name, for any other degree or diploma in any university or other tertiary institution without the prior approval of The University of Western Australia and where applicable, any partner institution responsible for the joint-award of this degree.

This thesis does not contain any material previously published or written by another person, except where due reference has been made in the text.

The work(s) are not in any way a violation or infringement of any copyright, trademark, patent, or other rights whatsoever of any person.

This thesis contains published work and/or work prepared for publication, some of which has been co-authored.

Signature:

Date: 21/12/2016

3

ABSTRACT

Plant growth, health and productivity are greatly affected by stress. In order for plants

to survive and thrive they must respond and adapt to these stresses at molecular,

cellular, physiological and biochemical levels. Understanding plant mitochondria is

critical for elucidating plant stress mechanisms as they are both an important

production site and target for reactive oxygen species. Although many years have

been spent on plant research there still remains a critical gap in our scientific

knowledge in regards to mitochondrial plant stress mechanisms. The post genomic

era has significantly altered the way in which research can be conducted and there

are now many new possibilities for investigating these previously unknown

mitochondrial plant stress mechanisms. UPOX1 has been identified as a protein that

is putatively targeted to the mitochondria and has been labelled as a highly stress

responsive hallmark of oxidative stress in Arabidopsis thaliana (Gadjev et al., 2006).

To uncover more details on this protein and to characterise UPOX1, a number of

different approaches were employed, including transcriptomic, proteomic and

bioinformatics studies. The profiles and kinetics of transcript induction were

determined for UPOX1 and associated genes including paralogs, putatively co-

expressed and co-regulated genes. It was found that the kinetics of UPOX1 induction

were unique, as they were significantly different from other UPOX homologs.

Therefore, this suggests the existence of independent regulatory mechanisms

between these homologs.

By investigating several putative cis-acting regulatory elements in the UPOX1

promoter, it was found that some of the stress-responsive pathways tested

overlapped for the induction of UPOX1 and other co-expressed genes. Furthermore,

through the analysis of defense signalling mutants, detailed insights were gained into

the intricate mechanisms underlying gene expression under stress conditions.

4

Transgenic plants with reduced or increased expression of UPOX1 were

characterised during normal and stress conditions. Finally, various proteomic

analyses were conducted to better understand the location of UPOX1. These

techniques included western blots, GFP tagging, in vitro uptake assays along with

alkaline extractions, PK digestions and BN-PAGE. These techniques confirmed that

UPOX1 is located in the mitochondrial intermembrane space and may also be

associated with the outer mitochondrial membrane.

Overall this research provides new detailed insights into this previously

uncharacterised marker gene of oxidative stress at both a transcriptomic and

proteomic level.

5

TABLE OF CONTENTS

FUNCTIONAL CHARACTERIZATION OF UPOX1, A MITOCHONDRIAL STRESS

RESPONSIVE PROTEIN IN ARABIDOPSIS THALIANA. ........................................................ 1

ABBREVIATIONS .................................................................................................................... 14

CHAPTER 1 .............................................................................................................................. 17

INTRODUCTION ....................................................................................................................... 17

1.1 RESEARCH IN THE POST GENOMIC ERA ..................................................................... 18

1.2 PLANT STRESS RESPONSE AND MANAGEMENT ....................................................... 20

1.3 TRANSCRIPTIONAL REGULATION ................................................................................. 22

1.4 ENDOSYMBIOTIC ORIGIN OF ORGANELLES ................................................................ 24

1.5 ORGANELLE BIOGENESIS .............................................................................................. 26

1.6 MITOCHONDRIAL STRUCTURE ...................................................................................... 30

1.7 MITOCHONDRIAL FUNCTION .......................................................................................... 31

1.7.1 The tricarboxylic acid cycle .................................................... 33

1.7.2 The electron transport chain .................................................. 34

1.8 PLANT MITOCHONDRIA ................................................................................................... 35

1.8.1 A functional Alternative Plant Respiratory Pathway ................... 36

1.8.2. Import of organelle proteins in eukaryotic cells ....................... 37

1.9 MITOCHONDRIAL STRESS RESPONSE ......................................................................... 38

1.10 CO-ORDINATED EXPRESSION OF NUCLEAR AND ORGANELLAR GENES ............ 40

1.10.1 Anterograde regulation ........................................................ 41

1.10.2 Retrograde regulation ......................................................... 42

1.10.3 Mitochondrial Retrograde Regulation ..................................... 42

1.10.4 Organelle communications between mitochondria and chloroplasts

.................................................................................................. 44

1.11 PROJECT AIMS AND APPROACHES ............................................................................ 45

CHAPTER 2 .............................................................................................................................. 51

MATERIALS AND METHODS ................................................................................................. 51

2.1 GENERAL CHEMICALS .................................................................................................... 52

2.2 ARABIDOPSIS GROWTH CONDITIONS .......................................................................... 52

2.2.1 Arabidopsis suspension cell culture ......................................... 52

2.2.2 Arabidopsis water culture ...................................................... 52

6

2.2.3 Arabidopsis plant material ..................................................... 52

2.2.4 Chemical treatment of Arabidopsis cell culture, water culture and

plant material ............................................................................... 53

2.2.5 UV treatment of Arabidopsis water culture ............................... 53

2.3 BACTERIAL TECHNIQUES ............................................................................................... 54

2.3.1 Bacterial cell strains .............................................................. 54

2.3.2 Transformation into JM109 and DH5 cells .............................. 54

2.3.3 Transformation into XL-Blue Super competent cells .................. 55

2.3.4 Transformation into XL-Gold Ultra competent cells .................... 55

2.4 NUCLEIC ACID MANIPULATION ...................................................................................... 55

2.4.1 Genomic DNA preparation ..................................................... 55

2.4.2 Total RNA isolation ............................................................... 56

2.4.3 Preparation of cDNA .............................................................. 57

2.4.4 Agarose gel electrophoresis ................................................... 58

2.4.5 Amplification of DNA by polymerase chain reaction ................... 58

2.4.6 TOPO-TA cloning into pCR2.1 ................................................. 58

2.4.7 Site Directed mutagenesis ..................................................... 59

2.4.8 Dpn1 Digestion .................................................................... 59

2.4.9 Restriction digests ................................................................ 60

2.4.10 Calf Intestinal Alkaline Phosphatase treatment ....................... 60

2.4.11 DNA ligation ....................................................................... 61

2.4.12 Plasmid Minipreps ............................................................... 61

2.4.13 Plasmid Midipreps ............................................................... 61

2.4.14 Plasmid Maxipreps .............................................................. 62

2.5 GENERATION OF ARABIDOPSIS MUTANT LINES ........................................................ 62

2.5.1 Preparation of Artificial microRNA constructs ............................ 63

2.5.2 Preparation of Agrobacterium tumefaciens electro-competent cells

.................................................................................................. 63

2.5.3 Transformation of electro-competent Agrobacterium ................. 63

2.5.4 Floral dipping ....................................................................... 64

2.5.5 Preparation of mutant lines .................................................... 64

2.6 TRANSIENT TRANSFORMATION OF ARABIDOPSIS CELL CULTURE AND LEAVES 65

2.6.1 Preparation of Arabidopsis suspension cells .............................. 65

2.6.2 Preparation of Arabidopsis leaves ........................................... 65

7

2.6.3 DNA precipitation onto gold microcarriers ................................ 65

2.6.4 Transformation of tissue ........................................................ 66

2.6.5 Reporter gene assays for luciferase and β-glucuronidase ........... 66

2.6.6 Normalisation of LUC and MUG activities ................................. 67

2.7 QUANTITATIVE RT-PCR ................................................................................................... 67

2.7.1 DNA standards ..................................................................... 67

2.7.2 Primer design and optimization .............................................. 67

2.7.3 qRT-PCR using the Roche Light Cycler® 480 ............................. 68

2.7.4 qRT-PCR Analysis ................................................................. 68

2.7.5 qRT-PCR Cross reactivity assays ............................................. 69

2.8 ORGANELLE AND PROTEIN ISOLATION METHODS .................................................... 69

2.8.1 Mitochondrial Isolation .......................................................... 69

2.8.2 Chloroplast Isolation ............................................................. 71

2.8.3 Protein translation ................................................................ 71

2.9 IN VIVO AND IN VITRO ASSAYS ...................................................................................... 72

2.9.1 In vivo GFP Assays ............................................................... 72

2.9.2 In vitro import of precursor proteins into mitochondria .............. 73

2.9.3 In vitro import of precursor proteins into pea chloroplasts ......... 74

2.9.4 Mitoplast preparation following mitochondrial in vitro imports .... 74

2.9.5 Alkaline extractions .............................................................. 75

2.9.6 Proteinase K titrations ........................................................... 76

2.9.7 SDS-PAGE ........................................................................... 76

2.10 WESTERN BLOTTING ..................................................................................................... 77

2.10.1 Blotting with nitrocellulose membranes.................................. 77

2.10.2 Blotting with PVDF membranes ............................................. 78

2.10.3 Immuno-detection .............................................................. 78

2.11.1 Preparation of BN PAGE gels ................................................ 79

2.11.2 BN PAGE sample preparation ................................................ 80

2.11.3 Wet Transfer of BN PAGE gels .............................................. 80

CHAPTER 3 .............................................................................................................................. 82

EXPRESSION AND REGULATION OF UPOX1 ...................................................................... 82

3.1 INTRODUCTION ................................................................................................................. 83

3.2 AIMS AND STRATEGIES .................................................................................................. 84

3.3 RESULTS ........................................................................................................................... 85

8

3.3.1 Comparisons between the UPOX1 family ................................. 85

3.3.2 Construction of a phylogenetic tree ......................................... 86

3.3.3 Establishment and optimisation of qRT-PCR assays ................... 86

3.3.4 Quantitative RT-PCR assays for UPOX1, UPOX2 and UPOX3 in

suspension cell culture .................................................................. 87

3.3.5 Transcript kinetics of nuclear genes encoding mitochondrial

proteins ....................................................................................... 89

3.3.6 Transcript analysis in response to UV stress ............................. 90

3.3.7 In silico response of UPOX1, UPOX2 and UPOX3 to a wide range of

stimuli and mutations .................................................................... 90

3.3.8 Mitochondrial genes induced under stress in signalling mutant

backgrounds ................................................................................ 91

3.3.9 GUS histochemical staining .................................................... 94

3.3.10 Identification of putative sequence elements in the upstream

region of UPOX1 ........................................................................... 94

3.3.11 Functional analysis of the B and I elements in the upstream

region of UPOX1 ........................................................................... 95

3.4 DISCUSSION ...................................................................................................................... 97

CHAPTER 4 ............................................................................................................................ 113

PROTEIN FUNCTION AND LOCALISATION ........................................................................ 113

4.1 INTRODUCTION ............................................................................................................... 114

4.2 AIMS AND STRATEGIES ................................................................................................ 115

4.3 RESULTS ......................................................................................................................... 116

4.3.1 Generation of UPOX1 under-expressing (knockdown) and over-

expression lines ........................................................................... 116

4.3.2 In vivo GFP ......................................................................... 118

4.3.3 In vitro mitochondrial imports ............................................... 119

4.3.4 In vitro chloroplast import .................................................... 120

4.3.5 Alkaline extractions ............................................................. 120

4.3.6 Proteinase K titrations into mitochondria and mitoplasts ........... 121

4.3.7 Blue Native PAGE ................................................................ 121

4.4 DISCUSSION .................................................................................................................... 122

CHAPTER 5 ............................................................................................................................ 133

GENERAL DISCUSSION ....................................................................................................... 133

5.1 SUMMARY OF AIMS AND APPROACHES .................................................................... 134

9

5.2 SUMMARY OF RESULTS ................................................................................................ 134

5.3 FUNCTIONAL IMPLICATIONS OF THE MITOCHONDRIAL STRESS RESPONSE ..... 136

5.4 RETROGRADE REGULATION OF NUCLEAR GENES ENCODING MITOCHONDRIAL

PROTEINS DURING STRESS AND THE MITOCHONDRIAL DYSFUNCTION STIMULON 139

REFERENCES ........................................................................................................................ 143

APPENDIX 1 ........................................................................................................................... 157

APPENDIX 2 ........................................................................................................................... 159

10

ACKNOWLEDGEMENTS

Coordinating Supervisor: Dr Olivier Van Aken

Co-supervisor: Professor James Whelan

Co-supervisor: Dr Monika Murcha

All experimental work and the initial thesis draft was carried out in the ARC Centre of

Excellence for Plant Energy Biology and the School of Chemistry and Biochemistry at

the University of Western Australia in Professor James Whelan’s lab.

This thesis would not have been possible without the help and guidance of several

individuals who contributed to the completion of this study.

I would like to thank my supervisor Prof Jim Whelan and for all his expertise, guidance

and for challenging me to do the best possible research throughout my PhD. My

sincere gratitude must go to Dr Olivier Van Aken for his encouragement, advice and

friendship throughout these years and supporting my thesis until the completion.

Thanks also to Dr Monika Murcha for her supervision of the proteomic studies and the

members of the Whelan lab for all the good times.

To my whole family, you are my whole world, you have made this life journey both

exciting and bearable. I know I always have you to count on and I’m so very grateful.

To Mali, Kasun’s Ammi, Thathi, Nisal and Sahan thank you for never giving up on me

and encouraging me to the finish line.

I especially want to thank my Ammi and Appachi, you have worked so hard and

sacrificed so much to give us every opportunity in life. You have always provided

unconditional love and care. I love you so much, I am so grateful for your unwavering

support and for all that you have given me. I would not have achieved anything in life

without your love and guidance.

To Kasun, my best friend, my soul-mate and my husband. There are no words to

convey how much I love you and how grateful I am to you for all that you have done to

11

support me through these years. The past several years have not been easy

academically and you have been my pillar of strength throughout, without you I could

never have achieved this. Thank you.

I dedicate this thesis to my parents and Kasun.

“When life is good do not take it for granted as it will pass. Be mindful, be

compassionate and nurture the circumstances that find you in this good time so it will

last longer. When life falls apart always remember that this too will pass. Life will have

its unexpected turns.”

-- Ajahn Brahm

12

AUTHORSHIP DECLARATION: CO-AUTHORED PUBLICATIONS

This thesis contains work that has been published.

Details of the work: Giraud E, Van Aken O, Uggalla V, Whelan J (2012) Redox Regulation of Mitochondrial

function in Plants; Plant, Cell & Environment 35(2): 271-280.

Location in thesis: Chapter 1&3

Student contribution to work: 10 %

Details of the work:

Van Aken O, Zhang B, Carrie C, Uggalla V, Paynter E, Giraud E and Whelan J (2009)

Defining the Mitochondrial Stress Response in Arabidopsis thaliana; Molecular Plant 2(6):

1310-1324.

Location in thesis: Chapter 4

Student contribution to work: 10 %

Details of the work: Ho LHM, Giraud E, Uggalla V, Lister R, Clifton R, Glen A, Thirkettle-Watts D, Van Aken O,

Whelan J (2008) Identification of regulatory pathways controlling gene expression of stress

responsive mitochondrial proteins in Arabidopsis; Plant Physiology 147(4): 1858-1873.

Location in thesis: Chapter 3

Student contribution to work: 10 %

Student signature:Date: 21/12/2016

13

I, Dr Olivier Van Aken certify that the student statements regarding their contribution to each of the works listed above are correct Coordinating supervisor signature: Date: 21-12-2016

Abbreviations

ABA abscisic acid

ABI4 abscisic acid insensitive 4

acetyl CoA acetyl coenzyme A

AOX alternative oxidase

At Arabidopsis thaliana

ATP adenosine tri-phosphate

BN-PAGE blue native polyacrylamide gel electrophoresis

CaMV cauliflower mosaic virus

CARE cis acting regulatory element

cDNA complementary deoxyribonucleic acid

CoA coenzyme A

Col-0 Columbia wild type

◦C degrees Celsius

DNA deoxyribonucleic acid

E.coli Escherichia coli

ETC electron transport chain

FAD flavin adenine dinucleotide

fmol femtomole

GC/MS Gas chromatography-mass spectroscopy

GUS β - glucuronidase

h hour

H2O2 hydrogen peroxide

HPLC high performance liquid chromatography

HR hypersensitive response

IM inner mitochondrial membrane

JA jasmonic acid

kb kilobase

kDa kilodalton

kPa kilopascal

L litre

LB Luria-Bertani

LUC luciferase

µg microgram

15

M molar

mA milliampere

MAPK mitogen activated protein kinases

Mb mega bases

MDS Mitochondrial Dysfunction Stimulon

mg milligram

min minutes

mL millilitre

mM millimolar

MnSOD Mn superoxide dismutase

mRNA messenger ribonucleic acid

MRR mitochondrial retrograde regulation

NAD(P)H nicotinamide adenine dinucleotide phosphate

NAD+ nicotinamide adenine dinucleotide (oxidised)

NADH nicotinamide adenine dinucleotide (reduced)

NADP-MDH NADP-Malate dehydrogenase

ng nanogram

NGEMP nuclear genes encoding mitochondrial proteins

OGDC 2-oxoglutarate dehydrogenase

OM outer mitochondrial membrane

PAGE polyacrylamide gel electrophoresis

PCD programmed cell death

PCR polymerase chain reaction

PD pyruvate dehydrogenase

PPR pentatricopeptide repeat

PVDF polyvinylidene fluoride

qRT-PCR quantitative real time polymerase chain reaction

redox reduction/oxidation

ROS reactive oxygen species

rpm revolutions per minute

s seconds

SA Salicylic acid

SAP shrimp alkaline phosphatase

SDS Sodium dodecyl sulphate

SDW sterile distilled water

TAE tris, acetic acid and EDTA

16

TBE tris, boric acid and EDTA

TCA tricarboxylic acid cycle

T-DNA transfer DNA

TIC translocases of the inner chloroplast envelope

TIM translocase of the inner mitochondrial membrane

TOC translocases of the outer chloroplast envelope

TOM translocases of the outer mitochondrial membrane

UPOX upregulated by oxidative stress

UQ ubiquinone

VDAC voltage dependent anion channel

[v/v] volume per volume

[w/v] weight per volume

Chapter 1

Introduction

18

1.1 Research in the Post Genomic Era

The complete sequencing of genomes has significantly altered the manner in which

research is conducted. In 1977 the bacteriophage φ X174 became the first

DNA-based genome to be completely sequenced (Sanger et al., 1977). The genomes

of more than 180 different organisms have been sequenced to date, with

representatives from all 5 biological kingdoms. In the year 2000 the genome for

Arabidopsis thaliana was completed making it the first plant genome to be sequenced

(The Arabidopsis Genome Initiative, 2000). Arabidopsis belongs to the Brassicaceae

family and over time it has become the pre-eminent plant model system. The reasons

as to why this model plant is extensively used in the field of molecular biology include

its relatively short life cycle of 6 weeks, small genome size of 125 Mb (Mega bases),

high progeny yield of up to 10 000 seeds per plant and the availability of more than 30

000 gene knockout lines. Consequently, there are large databases with information

pertaining to its growth and development (Lister et al., 2009). These factors in

combination with the high quality genome information have allowed for great progress

in both forward and reverse genetic research. The technological advances in the fields

of transcriptomics, proteomics and metabolomics that have arisen in the post genomic

era have now enabled research to be conducted at an unprecedented scale. It is

possible to gain significant insight into the complex physiological processes by

analysis at any of these 'omic' levels, and combinatorial approaches can be in

particular very powerful.

Transcriptomics is the study of the abundance of all transcripts in an organism. The

transcriptome is dynamic, as it is the sum of synthesis and degradation of thousands

of mRNAs in a cell (Meyuhas and Perry, 1979; Wilhelm et al., 2008). These changes

can be due to changes in the organism’s development or in response to external

stimuli (Holtorf et al., 2002). Microarrays investigating gene expression are a popular

method used to study whole genome transcriptomes. The basic principle behind

19

microarrays (e.g. Affymetrix GeneChip microarrays) involves the hybridisation of

fluorescently labelled cDNA (complementary deoxyribonucleic acid) to complementary

cDNA probes immobilised on small glass platforms or chips. The probes are designed

using genome sequence information and their position on the chip is catalogued. The

fluorescently labelled targets comprising cRNA or cDNA from individually prepared

samples can originate from for instance mutant lines, responses to external stimuli or

particular developmental stage (Ruan et al., 1998). The quantitative fluorescence

image can be monitored using a high resolution laser scanner from which the

transcript abundance can be inferred (Holtorf et al., 2002). A major limitation in the

use of microarrays is the requirement for well-annotated transcript information to

design the relevant probes. The development of high-throughput next generation

sequencing technologies allows for relatively rapid sequencing of the entire

transcriptome, circumventing the need for a priori genome information, and such

‘deep sequencing’ approaches are now more widespread and cost-effective (Wang et

al., 2012).

Proteomics is the study of the entire complement of proteins within an organism, cell

or tissue type (Chen and Harmon, 2006). Research using proteomic methods

significantly differ from that of transcriptomics. Transcriptomics can be performed

using a very small amount of mRNA sample by amplification of the starting material

via reverse transcription and PCR amplification. The main limitation to proteomics is

the large amount of starting material required to perform proteomic analysis. Another

limitation to proteomic studies is the efficiency with which proteins can be extracted

from tissues. However, over the last decade this area of research has been improving

at an increasing rate. These methods can evaluate small fragments of peptides using

tandem mass spectrometry (MS/MS) and several methods that involve affinity based

pull down assays using intact proteins to identify interactions such as Tandem affinity

purification coupled to MS/MS (ref). Protein complexes can be separated in their

20

native configurations using Blue Native PAGE analysis (BN-PAGE) (Cannon and

Webb-Robertson, 2006).

Metabolomics is potentially a very powerful ‘omic’ technique. Unlike transcriptomics

and proteomics this method of research doesn’t require a fully sequenced genome,

however, it does require a comprehensive database on which to cross check

molecular profiles against previously identified compounds. Until these databases

become sufficiently comprehensive, many questions pertaining to the metabolic

activities of organisms will remain unanswered (Fukushima et al., 2014). This method

involves extracting chemical constituents from cells and then identifying these

extracted fractions using techniques such as gas chromatography-mass spectrometry

(GC/MS) and high performance liquid chromatography (HPLC) (Roessner et al., 2001;

Holtorf et al., 2002).

To maintain the pace of research and to elucidate more in-depth mechanisms of

stress response and organelle biogenesis it is now required that multiple ‘omic’ levels

be studied in collaboration with each other.

1.2 Plant Stress Response and Management

All organisms are exposed to stress and have developed responses and adaptations

in order to survive. Plant stress has been defined as the conditions in which the

environment adversely affects plant growth and performance (Millar et al., 2001).

Stress can involve a constraint or a highly unpredictable fluctuation in the regular

metabolic patterns that usually cause injury and disease (Gaspar et al., 2002).

There are two types of stresses that may affect plants, abiotic and biotic. Abiotic

stress is caused by environmental factors such as drought, extreme temperatures,

soil, mineral deficiencies, salinity, chilling, hypoxia and high winds (Bartels and

Dinakar, 2013; Benikhlef et al., 2013; Jin et al., 2013; Mustroph et al., 2014;

Nakashima et al., 2014; Fernandes et al., 2016). Biotic stress is caused by pathogenic

21

organisms and includes viral, bacterial and fungal infections (Bahar et al., 2016; Misra

et al., 2016). To manage stress effectively the plant must firstly recognise the stress

imposed on it, activate stress response pathways and then respond to the stress

effectively.

Recognition of abiotic and biotic stresses affecting plants is required at an early stage

in order for plants to be resilient and resistant to stresses. A hypersensitive response

is commonly exhibited by plants in defense against pathogen invasion. This response

involves the formation of localised regions of cell death in order to limit cell transfer of

the pathogen (Heath, 2000).

The understanding of abiotic and biotic stress response in plants is an important and

challenging topic. The availability of the complete genome sequence of Arabidopsis

thaliana has facilitated access to physiological and molecular biological analysis.

Stress tolerance mechanisms include transcriptional changes in genes, in particular

stress responsive genes. The products of such genes can be grouped into two

categories; products that can directly protect plants from stresses and products that

produce a stress response by altering gene expression and/or signal transduction

pathways (Shinozaki et al., 2003; Teige et al., 2004). The first group includes

detoxification enzymes and osmo-protectants, both of which can directly protect plants

on a cellular level. Products from the second group include transcription factors and

mitogen activated protein kinases (MAPK).

A single stress condition can activate numerous signalling pathways, these pathways

may interact with one another and form intertwined response networks. Plants have

evolved complicated and sophisticated systems in response to complex stresses that

can affect growth, development and altered responses to endogenous and

environmental cues (De Caluwé et al., 2016). Salicylic acid (SA), abscisic acid (ABA),

jasmonic acid (JA) and ethylene are all hormones involved in plant stress signalling

pathways. SA accumulates following viral, fungal and bacterial pathogen infections

22

(White, 1979; Sticher et al., 1997; Huang et al., 2003) and results in the induction of

pathogen-related (PR) genes. SA has been shown to be involved in systemic acquired

resistance (SAR), in which a pathogen attack in one part of a plant can result in

pathogen resistance in non-infected areas (Vlot et al., 2009). Osmotic stress activates

the biosynthesis of ABA through the regulation of several biosynthetic proteins in the

plastid resulting in the accumulation of ABA within the cell (Skriver and Mundy, 1990).

ABA is produced in the roots of the plant under low water conditions and then

transported to the leaves where it reduces water loss e.g. by stimulating stomatal

closure (Schachtman and Goodger 2008). Wounding and specific fungal elicitors

increase the biosynthesis of JA and thus activate expression of proteins involved in

plant defense (Koo et al., 2009). The majority of the regulation following hormonal

stimuli occurs at the level of gene expression. Gene expression is a broad term,

defined as transcription and translation of a gene to produce a protein. There are

many possible levels of regulation i.e. transcriptional, post-transcriptional, translational

and post-translational, which together will lead to controlled levels of mRNA and

protein abundance

(Van Aken et al., 2016).

1.3 Transcriptional Regulation

In eukaryotes, transcriptional regulation primarily occurs at the level of transcriptional

initiation. This is facilitated by cis acting regulatory element (CAREs) within the

promoter areas of genes. The CAREs are recognised and bound by transcription

factors that recruit basal transcriptional machinery or chromatin remodelling

complexes (Gentry and Hennig, 2014; Han et al., 2015). These transcription factors

usually contain several domains including a DNA binding domain, oligomerisation site,

transcriptional regulation domain and nuclear localisation signal. Highly conserved

amino acids (that tend to be basic) determine the specificity of interactions within the

23

DNA binding site. The oligomerisation site is the site at which homo or hetero

oligomers are formed. The transcriptional regulation domain is the site of repression

or activation depending on the transcription factor thus allowing for inhibition or

excitation of a gene (Yanagisawa and Sheen, 1998; Liu et al., 1999). At each of these

sites there is the possibility of further specificity depending on the organism and the

complexity of its transcriptional regulation mechanisms.

A co-ordinated and dynamic response to plant stress is required as the mechanism of

plant stress response is highly complex. Most genes are induced in a stimulus specific

manner, however stress responsive genes tend to display complex overlapping

patterns of response to some common biotic and abiotic stress conditions (Chen et

al., 2002; Weltmeier et al., 2009). The signalling pathways within Arabidopsis plants

following cold, drought and high salinity conditions showed a higher degree of overlap

than the signalling pathways following drought and heat stress conditions (Chow et

al., 2014). An overlap between different stress conditions is also evident in

transcriptomic profiling studies as shown by Gadjev et al., 2006; Gupta et al., 2016.

The large networks of transcription factors are the key to the plant genomes’ ability to

modulate its response to fluctuating environmental conditions. It was estimated that

the Arabidopsis genome encodes greater than 2000 transcription factors, however

less than 100 have available experimentally confirmed regulatory interactions (Barah

et al., 2015). Stress inducible transcription factors include the WRKY family, which is

a transcription factor family unique to plants involved in biotic, abiotic stresses, as well

as plant development (Rushton et al., 2010). Another example are the ethylene-

responsive element binding factor (ERF) family which is one of the third largest

transcription factor gene families in Arabidopsis (Singh et al., 2002; Shinozaki et al.,

2003). These transcription factors have been shown to work independently and

co-operatively to coordinate appropriate stress responses (Shinozaki et al., 2003).

24

Eukaryotic transcriptional regulation tends to be combinatorial and involves

interactions between several transcription factors that bind to the same or different

CAREs (Elkon et al., 2003). Studies have shown that transcription factors are over

represented in large genomes. This also correlates with an organism’s ability to adapt

and inhabit environmental conditions that are prone to change. Therefore it can be

concluded that having a larger genome and combinatorial transcriptional regulation

mechanisms is advantageous for the survival of multicellular organisms (Elkon et al.,

2003; Wray et al., 2003).

1.4 Endosymbiotic Origin of Organelles

A defining characteristic of eukaryotic cells is the presence of compartmentalised

specialised organelles. The formation of eukaryotic cells is hypothesised to have

occurred as a result of endosymbiotic events (Hedges et al., 2001). The

endosymbiotic origin was first hypothesised in the late 19th century however it was

largely ignored. It was only after the presence of overwhelming evidence for

endosymbiosis that the theory became widely accepted. The discovery of organellar

deoxyribonucleic acid (DNA) was a key element for the theory of endosymbiotic origin

of organelles. This is because the DNA within the mitochondrial and plastids differs

from that of the DNA within the nucleus, and it resembles the circular shaped DNA

found in bacteria. Further evidence for the endosymbiotic origin of organelles includes

the process of formation of mitochondria and plastids, as it involves a similar process

to binary fission. The proteins of bacteria and organelles both use N-formylmethionine

as an initiating amino acid and mitochondria have enzymes and transport systems

similar to that of bacteria. The ribosomes are similar to the 70S ribosomes found in

bacteria. Cyanobacteria have similar structures to the thylakoids and chlorophylls of

the chloroplasts and mitochondria and plastids are of similar size to bacteria. Finally

most of the genes needed by mitochondria and plastids are produced within the

25

nucleus and this is consistent with the theory that following endosymbiosis there is an

increased dependence of the eukaryotic host (Gray, 1989; van der Giezen et al.,

2005; de Duve, 2007; Poole and Penny, 2007).

This wide array of evidence also suggests that there were two symbiotic events in the

history of the eukaryotic cell; one pre-mitochondrial and a later event after

mitochondria were already assimilated (Hedges et al., 2001). The first endosymbiotic

event is estimated to have occurred 2 billion years ago between an anaerobic pro-

eukaryote that was dependent on glycolysis for energy production, and an α-

proteobacteria probably related to Rickettsia (Emelyanov, 2001). It is hypothesised

that at the time of this endosymbiotic event the atmospheric oxygen levels were

relatively low and the endosymbiont produced (Adenosine Tri-Phosphate) ATP and

hydrogen (Martin and Müller, 1998). The theory of endosymbiosis is also used to

explain the tendency of eukaryotic genes to display an archaeal or bacterial ancestry.

For example genes involved in processes such as transcription and translation tend to

be closely related to archaeal genes. However genes involved in cellular metabolic

processes such as lipid biosynthesis tend to be closely related to eubacterial

(Villanueva et al., 2016).

Approximately 1.5 billion years ago the second endosymbiotic event is hypothesised

to have taken place. The endosymbiotic event was believed to have occurred

between a eukaryote possessing mitochondria and a cyanobacterial ancestor that

was capable of photosynthesis. This resulted in the formation of plastids, leading to a

distinct lineage of eukaryotes, the forebears of modern plants (Palenik, 2002).

There still remains debate over the driving force behind the endosymbiotic events.

Current theories focus on the functions of the endosymbiont: hydrogen production,

and methane consumption are proposed by (Kim et al., 2014; Paranjape and

Hallenbeck, 2015). Additionally, the hypothesis of ‘ox-tox’ is also suggested by

26

(Stefano and Kream, 2016) that centres around the increasing oxygen levels and the

adaptations required for survival under these changing conditions.

1.5 Organelle Biogenesis

The transition from endosymbiont to a eukaryotic cell containing organelles resulted in

the formation of very diverse but distinct sets of enzymes, whose

compartmentalisation has allowed for a level of specialisation unachievable in

prokaryotes (Aitchison et al., 1992). The active proliferation and differentiation of

these organelles, that resulted in an increase in organelle number and mass, was

termed organelle biogenesis (Nisoli et al., 2004). An important event following

endosymbiosis was the vast genome reduction of the endosymbiont that took place

within the newly formed eukaryotic cells. The magnitude of the organelle genome

reduction can be fully comprehended when comparing the number of protein encoding

genes between -proteobacteria, modern day cyanobacteria and mitochondria. An -

proteobacterial genome can contain up to 9000 protein coding genes (Ettema and

Andersson, 2009), a cyanobacterium contains greater than 2000 protein coding genes

(Fujisawa et al., 2014) and in contrast, modern day mitochondria in P. falciparum and

R. americana respectively contain only 3 and 67 protein encoding genes respectively

(Gray et al., 1999).

Many organelle genes have been functionally transferred to the nucleus or have been

replaced by nuclear genes of similar function (Adams et al., 2002; Bergthorsson et al.,

2003). A clear example of this is demonstrated by the fact that there are over 2000

gene products forming the mitochondrial proteome however only 57 are encoded by

the mitochondrial genome in Arabidopsis thaliana (Unseld et al., 1997; Sato et al.,

1999). Despite the extensive reductions, the genetic function of organelle genomes

27

still remains well conserved. This vast reduction in the organelle genome necessitated

the evolution of mechanisms enabling organelle biogenesis. This included metabolite

exchange pathways, protein targeting and translocation systems (Dyall et al., 2004).

Considering the majority of proteins within mitochondria and chloroplasts require

importation from nuclear encoded cytosolicly synthesised precursors, organelles

require multi-subunit translocons and protein import machinery to facilitate this

process. In excess of 2000 proteins are imported into each organelle and although

there are similarities between the basics of the import apparatus of each organelle

they remain very specialised (Kleffmann et al., 2004; Millar, 2007). Examples of their

similarities include the presence of multi subunit translocons required to facilitate

protein import into the organelles. There are translocases of the outer chloroplast

envelope and a translocases of the outer mitochondria membrane (TOC and TOM)

respectively and there are also a translocases of the inner chloroplast envelope and

mitochondrial membrane (TIC and TIM) (Kleffmann et al., 2004; Millar, 2007). The

TOC and TOM complexes both have central components that precursor-proteins

dock to in order to mediate protein transfer, these are both cation selective channels

and the initial sorting of proteins occurs here. However the mode of translocation of

proteins differ between the two organelles; in mitochondria the passage of pre-

proteins is driven by their increasing affinity for the translocation subunits and within

chloroplasts GTP-binding and hydrolysis by the receptors are required for

translocation(Schleiff and Becker, 2010). Generally proteins are imported into

mitochondria and chloroplasts post translationally and several protein machineries

mediate this transport. Both the TOM and TOC complexes consist of receptors as

TOM20, TOM70, TOM22, TOC34, TOC64 and TOC159, furthermore there are pore

forming proteins such as TOM40 and TOC75. In addition there are complexes

involved in the assembly of additional TOM and TOC complexes, in the stabilisation of

28

these complexes and also the transfer of precursor proteins between complexes. In

plants there are isoforms of these TOMs and TOCs (Schleiff and Becker, 2010).

Within the chloroplasts there is a Toc-Tic supercomplex and Tic110, Tic22 and Tic20

are three inner membrane proteins that are associated with this supercomplex. They

assist in forming functional import sites on this supercomplex. To date it appears that

the assembly of a functional inner membrane translocon appears to be mediated by

associations with the Toc complex as there is no evidence to state that a stable

complex consisting only of Tic components exists (Soll and Schleiff, 2004). Similarly

to the Tic complexes there are several Tim complexes. These facilitate the

translocation of proteins across the inner membrane of the mitochondria. The main

Tim complexes include Tim23, Tim22 with the main subunits Tim17 and Tim 23. The

exact roles differ somewhat between these complexes as Tim23 facilitates the

translocation of proteins targeted to the matrix whereas Tim22 facilitates the

integration of carrier pre-proteins into the inner membrane of mitochondria. However

in general all the Tim complexes do allow for import of proteins into mitochondria

(Sirrenberg et al., 1996; Paschen et al., 2000). These similarities between Tic and

Tims and Tocs and Toms give rise to the concept that these organelles have co-

evolved, possibly explaining the economy of function via protein dual targeting (Carrie

et al., 2009).

There is debate over the method of biogenesis of mitochondria within the cell. It has

been hypothesised new mitochondria are derived via the fission of existing organelles

and alternatively it has also been hypothesised that the method of biogenesis of

mitochondria involves maturation of precursor organelles termed pro-mitochondria.

The pro mitochondria are more of a rest and storage state for instance during seed

production or spore formation etc. that needs to be resurrected when the seeds are

imbibed (Law et al., 2014). To date there is significant evidence for both models of

biogenesis.

29

Evidence for the first model involving fission is supported by the fact that mitochondria

and plastids both contain remnants of their ancestral genomes. These remnants are

essential components of these organelles and therefore these organelles are

incapable of arising in cells de novo. This contrasts against other organelles such as

the nuclei, peroxisomes and endoplasmic reticulum that have been observed to arise

de novo in cells (Dimitrov et al., 2013).

The complex machinery involved in the cell division of mitochondria and chloroplasts

include both host derived proteins and orthologs from their associated bacterial

ancestors(Yang et al., 2008). There are distinctions between the protein complexes

involved in both mitochondrial and chloroplastic fission however they function in the

same manner by a synchronized constriction of the outer and inner membranes in

order to produce two daughter cells (Osteryoung and Nunnari, 2003). There have

been significant studies examining mitochondrial populations in yeast and in

mammalian systems. These studies have accumulated further evidence to support the

growth and division model of mitochondrial biogenesis (Nisoli et al., 2004). Generally,

mitochondrial fission has been shown to coincide with mitosis with mitochondrial mass

increasing from the beginning of the S-phase until M-phase and results in each

daughter cell receiving a roughly equal complement of mitochondria(Sänger et al.,

2000). Although this is what is known to generally occur it has also been observed in

mammalian muscles that mitochondrial proliferation occurred in response to an

increase in physical activity (Brunk, 1981).

The second model for mitochondrial biogenesis centres on pro-mitochondria. There is

still no conclusive analysis outlining the origin of pro-mitochondria however it is

believed that they are caused by the degeneration of mature mitochondria (Day et al.,

2004). These pro-mitochondria are thought to be simple unstructured organelles

(Plattner et al., 1970). The evidence for this model was observed in yeast cells when

they transitioned from anaerobic to aerobic growth and also during seed germination

30

(Rosenfeld et al., 2004; Howell et al., 2006). In both these situations it appears that

the maturation model is applicable for cells undergoing a fundamental phase-

transition.

It has been shown under electron microscopy that during the phase transition energy

intensive period of seed germination the presence of poorly differentiated

mitochondria lacking cristae and matrix are evident. These pro mitochondria were

observed to have only a fraction of the usual protein complement and although they

possessed the protein machinery for import they were not capable of importing

proteins (Howell et al., 2006, 2007). Continuing observations taken 30 min following

the imbibition showed significantly increased rates of protein import. Thus it has been

proposed that during these fundamental phase changes such as during the

germination process, pro-mitochondria mature into metabolically active mitochondria,

which subsequently give rise to more mitochondria via growth and division (Howell et

al., 2006, 2007).

1.6 Mitochondrial Structure

Mitochondria are bioenergetic organelles, they display variations in form, however

generally vary in size between 1 µm to 2 µm in diameter (Siedow and Umbach, 1995).

Mitochondria contain four separate compartments, the outer membrane, the inner

membrane, the inter-membrane space and the mitochondrial matrix as shown in

Figure 1.1.The outer and inner membranes enclose a dense matrix incorporating

enzymes and multiple copies of a genome that encode a small number of inner

membrane proteins and the RNAs required for their translation (Frey and Mannella,

2000). The majority of mitochondrial proteins are nuclear encoded and imported from

the cytosol. The outer mitochondrial membrane (OM) is impermeable to large proteins

greater than 10kDa yet permeable to solutes, whilst the inner mitochondrial

membrane (IM) is osmotically active and impermeable to most solutes. A vital protein

31

required for import is Voltage Dependent Anion Chanel (VDAC) also known as Porin

and allows free passage to small molecules including NAD(P)H (Frey et al., 2002).

The IM contains most of the components required for the electron transport chain

(ETC) in addition to several other mitochondrial transporters. The shape of the

mitochondrial IM changes in response to metabolic and osmotic conditions. Oxidative

phosphorylation requires the rapid diffusion of ions and substrates across the IM.

Therefore the morphology of the cristae may regulate the rate of movement of ions

and substrates through the IM (Frey and Mannella, 2000). The intermembrane space

contains cytochrome c also essential for the ETC. The mitochondrial matrix is the

location where most of the tricarboxylic acid cycle (TCA) takes place, in addition to

various other metabolic processes such as amino acid metabolism and glycine

oxidation (Bowsher and Tobin, 2001; Rasmusson et al., 2004). Under normal

conditions the matrix is not electron dense and the matrix is expanded, however the

intra cristae spaces can become dilated and the matrix can become electron dense

and therefore this is part of a feedback mechanism that allows mitochondria to

respond to environmental perturbations (Frey and Mannella, 2000).

1.7 Mitochondrial Function

Mitochondria are multifunctional and semi-autonomous organelles. They carry out a

variety of functions, of which their primary roles are the oxidation of organic acids via

the tricarboxylic acid cycle and the synthesis of ATP coupled to the transfer of

electrons from NADH to oxygen via the electron transport chain.

Mitochondria function in several different ways however the most prominent of them is

its role to produce ATP (by the phosphorylation of ADP) through respiration. In

addition to the TCA cycle mitochondria play a crucial role in plant defense

mechanisms. The hypersensitive response (HR) and programmed cell death (PCD)

32

are both influenced by mitochondrial function (Robson and Vanlerberghe, 2002;

Vacca et al., 2004).

There is evidence supporting a leading role for mitochondria in PCD involving

components of the mETC, FoF1 ATP synthase, cardiolipins, and ATPase AtOM66

(Zhang et al., 2014; Van Aken and Van Breusegem, 2015). Furthermore, it is

hypothesised that both the mitochondria and chloroplast both play an important role

during plant PCD. This is thought to occur by changes in redox levels and energy

states or metabolic equilibriums producing large quantities of ROS or the release of

pro-death factors which are yet to be fully understood. In addition to the mitochondria

other organelles that contribute to PCD include the vacuole (e.g., via vacuolar

processing enzymes and autophagy) and the ER (Van Aken and Van Breusegem,

2015).

VDAC is a large protein located on the outer mitochondrial membrane is known to be

an important component involved in PCD and HR. In Arabidopsis, proteins known as

hairpins that are produced in nature by certain bacterial plant pathogens were shown

to inactivate mitochondria in suspension cells. These cells were shown to have altered

mitochondrial functions. These were recognized by the enhanced generation of

mitochondrial reactive oxygen species and nitric oxide. In addition it was also evident

that there was a decrease of the mitochondrial membrane potential and of ATP

production, and the induced-expression of alternative oxidase gene and small heat

shock protein genes (Krause and Durner, 2004; Kusano et al., 2009). Further

evidence of this includes the fact that alternative oxidase (AOX) is known to be a

critical component involved in the HR salicylic hydroxamic acid-sensitive pathway

which is important in viral resistance (Ordog et al., 2002). The overexpression of AOX

in transgenic tobacco (Nicotiana tabacum) plants resulted in smaller hypersensitive

response lesions; this suggests a strong link is present between mitochondrial

function and programmed cell death. Furthermore VDAC a large protein located on

33

the outer mitochondrial membrane is also known to be an important component

involved in PCD and HR.

Mitochondria are also involved in a dynamic relationship with other cellular organelles

such as chloroplasts and peroxisomes to facilitate the efficient functioning of cells.

This can be evidenced during photorespiration, within photosynthetic cells a major

demand on mitochondria is the oxidation of glycine to serine generated during

photorespiration (Raghavendra and Padmasree, 2003; Balmer et al., 2004; Wong et

al., 2004). Furthermore mitochondria are able to sense and responds to

environmental changes and variations in metabolic demands of the other organelles,

for example mitochondrial and chloroplastic reductant export is suggested to co-exist

with peroxisomal and mitochondrial reductant import (Foyer and Noctor, 2005).

Mitochondria also play crucial roles in a number of essential biosynthetic processes

such as nitrogen fixation, the biosynthesis of amino acids and vitamin cofactors

(Mackenzie and McIntosh, 1999). For example plant mitochondria have been shown

to be the site of fatty acid synthesis and enzymes involved in the synthesis of cystein,

glycine and proline have been identified in mitochondria (Taylor et al., 2004).

1.7.1 The tricarboxylic acid cycle

The TCA cycle is the second stage of cellular respiration. Prior to the TCA cycle

glucose can be metabolised to pyruvate anaerobically as shown in Figure 1.2 A, this

however only synthesises a fraction of the ATP available from glucose. The aerobic

processing of glucose begins with the complete oxidation of glucose derivatives to

carbon dioxide. This oxidation takes place in the TCA cycle that is comprised of a

series of reactions also known as the citric acid cycle or the Krebs cycle. This

metabolic process occurs in most plants, animals, fungi and many bacteria. In all

organisms except bacteria the TCA cycle is carried out predominantly in the matrix of

the mitochondria. The larger molecules such as glucose, other sugars, fatty acids and

34

amino acids enter the TCA cycle after conversion to the break-down product acetyl

coenzyme A (acetyl CoA). The TCA cycle has 8 main steps as shown in Figure 1.2 B.

The first step is initiated by the reaction of acetyl CoA and oxaloacetate, this forms

citrate and produces the release of coenzyme A (CoA-SH). Following this, a

succession of reactions occur, citrate is rearranged to form isocitrate as shown in step

2 of Figure 1.2 B; isocitrate then loses a molecule of carbon dioxide and undergoes

oxidation to form alpha-ketoglutarate in step 3; alpha-ketoglutarate loses a molecule

of carbon dioxide and is oxidized to form succinyl CoA in step 4; in step 5 succinyl

CoA is enzymatically converted to succinate; succinate is then oxidized to fumarate in

step 6; in step 7 fumarate is hydrated to produce malate; and finally in step 8 malate is

oxidized to oxaloacetate. Each complete turn of the cycle results in the regeneration

of oxaloacetate and the formation of two molecules of carbon dioxide(Martin and

Vagelos, 1962).

Energy is produced and relayed through the many Redox (reduction/oxidation)

reactions and following several steps of the TCA cycle. One molecule of ATP is

produced following step 5 however a large proportion of energy is captured by

nicotinamide adenine dinucleotide (NAD+) and flavin adenine dinucleotide (FAD) that

are later converted to ATP. For each cycle three molecules of NAD+ are reduced to

NADH and one molecule of FAD is reduced to FADH2. The energy within these

molecules are then transfered to the electron transport chain, a pathway that is part

of the third stage of cellular respiration. The electron transport chain in turn releases

energy so that it can be converted to ATP through the process of oxidative

phosphorylation(Martin and Vagelos, 1962; Carrari et al., 2003; Pollard, 2003).

1.7.2 The electron transport chain

The ETC comprises several multi-subunit complexes and the redox energy from

NADH and FADH2 is transferred to oxygen via several steps as shown in Figure 1.1.

35

Complex I is a NADH-ubiquinone oxidoreductase, it catalyses the oxidation of NADH

and reduction of ubiquinone (UQ) a mobile redox carrier. Complex I comprise over 30

subunits in Arabidopsis however only 9 of them are encoded by the mitochondrial

genome. Complex II in Arabidopsis is composed of 8 nuclear encoded subunits and is

responsible for catalysing the transfer of electrons from succinate to UQ via covalently

bound FAD (Eubel et al., 2003). Complex III is also known as the bc1 complex, it is a

UQ H2-ferrocytochrome c oxidoreductase. Complex III then catalyses the transfer of

electrons from UQ to cytochrome c. This process involves the Rieske protein an (iron

sulfur center) and cytochrome b. Complex IV catalyses the oxidation of cytochrome c

and the reduction of O2 to water. This process is efficient however a small percentage

of electrons may prematurely reduce oxygen, this forms reactive oxygen species such

as superoxide. Superoxides can cause oxidative stress and damage. The transfer of

electrons through these complexes is coupled to the translocation of protons from the

matrix into the intermembrane space. As the proton gradient increases a strong

electrochemical gradient is formed across the inner membrane. This proton gradient

drives the synthesis of ATP from ADP and inorganic phosphate at Complex V which is

also refered to as ATP synthase, this process is called chemiosmosis (Mitchell and

Moyle, 1967).

1.8 Plant Mitochondria

Over the last 50-60 years researchers have been investigating the mitochondria of

plants and we now know there is a remarkable variation in the functional abilities of

this organelle relative to their mammalian and other non-photosynthetic counterparts.

In order for plants to survive as sessile organisms, plant mitochondria have evolved

unique strategies to enhance their survival capabilities. Mitochondrial genomes

encode a very small proportion of the genetic information required for their function;

36

the majority of the information is nuclear derived. This is hypothesised to have been a

result of nuclear-mitochondrial co-evolution within the plant kingdom.

1.8.1 A functional Alternative Plant Respiratory Pathway

As a unique advantage for survival, plants, some protists, fungi and algae possess a

(second) alternative respiration pathway as shown in Figure 1.1.These alternative

pathways bypass energy conservation by circumventing the utilization of the

electrochemical proton gradient. Components of this pathway include a type II

NAD(P)H dehydrogenases (NDHs), which bypasses proton-pumping complex I or

allows oxidation of cytoplasmic NAD(P)H; the alternative oxidase (AOX), which

bypasses proton -pumping complexes III and IV; and uncoupling proteins (UCP), that

bypass the ATP synthase by directly dissipating the proton gradient (Escobar et al.,

2004; Rasmusson et al., 2004).

The terminal oxidase in the alternative pathway is AOX. AOX is a nuclear encoded

mitochondrial protein of approximately 34 kDa. AOX is bound to the membrane bilayer

of mitochondria; it contains a coupled bi-nuclear iron centre with histidines and four

glutamate residues. It is a member of the di-iron carboxylate group that is

distinguishable by the presence of six conserved amino acids proposed to represent

iron-binding residues(Berthold et al., 2000).

The alternative respiratory pathway allows for a greater level of flexibility for regulating

the redox state of the mitochondrial and cytoplasmic matrix NAD(P)H pools as neither

of the AOX or NDHs contribute to proton pumping or ATP synthesis and bypass

adenylate control (Rasmusson et al., 2004). The level of ubiquinone can be altered by

the action of these alternative respiratory proteins consequently the level of reactive

oxygen species (ROS) can also be modified. There have been further observations

that show the enzymes of the alternative respiratory pathway play key roles in plant

stress response (Moore et al., 2002). In addition, findings also show that these

37

alternative respiratory proteins can also alter cellular redox, energy levels and can

minimise the production of ROS (van Lis and Atteia, 2004). Therefore mitochondria in

plants are both a target of oxidative damage and agents involved in stress response

and detoxification (Van Aken et al., 2009).

1.8.2. Import of organelle proteins in eukaryotic cells

The mitochondrial import apparatus is essential for the functioning of all eukaryotic

cells as the majority of more than 1000 proteins within the mitochondria are nuclear-

encoded and imported (Murcha et al., 2014). The process of discrimination and

sorting of nuclear encoded mitochondrial targeted proteins falls on the import

mechanisms. Although the Arabidopsis genome was sequenced many years ago

(AGI, 2000) the components and mechanisms involved in Arabidopsis protein import

continue to be studied in further detail. While the yeast model system doesn’t reflect

the complexity of higher plants there are several components of the protein import

machinery that have been conserved between these species. As the import

mechanisms in yeast have been studied more intensively this assists in the study of

the components involved in the import machinery within Arabidopsis.

There are a variety of mitochondrial import pathways that operate in eukaryotic

organisms. First, the general import pathway imports mitochondrial proteins with N-

terminal targeting signals. This pathways uses the translocase of the outer membrane

(TOM) and the translocases of the inner membrane 17:23 (TIM17:23). For these

proteins the primary signal defines the matrix location with secondary signals sorting

proteins into the inner membrane or intermembrane space (IMS) (Murcha et al.,

2014).

The mitochondrial carrier import pathway involves the import of inner membrane

proteins that contain multiple transmembrane regions. This pathway varies from the

38

general pathway as it utilises the TOM complex, a variety of small TIM proteins in the

intermembrane space and a translocase of the inner membrane 22 (TIM 22). Most of

these proteins’ targeting signals are located in the mature region of the protein rather

than the N-terminal region (Duncan et al., 2013).

The import of IMS proteins involves another protein import pathway. The signals on

these proteins are also in the mature protein and this pathway involves twin cysteine

residues and utilizes the TOM complex and the mitochondrial IMS assembly pathway

(MIA) machinery. The fourth import pathway involves the import of β-barrel proteins

into the outer membrane. This pathway uses a TOM and a sorting and assembly

machinery (SAM). In addition to these two outer membrane complexes small TIM

proteins from the IMS are also involved to import these β-barrel proteins (Murcha et

al., 2014). There are several additional specialised protein import pathways that are

known to exist for specific protein groups/proteins however to date these are not fully

understood (Bauer et al., 2015).

1.9 Mitochondrial Stress Response

As plants are continuously challenged by abiotic and biotic stresses it is vital that they

have mechanisms to counteract the damage caused by these external stresses. To

deal with stress, plants have several phyto-hormone-based pathways that signal

stress responses that crucially allow for stress tolerance (Bostock, 2005). These

stress responses include modification of the transcriptome and various post-

transcriptional changes (Koornneef and Pieterse, 2008). Mitochondria are a target of

oxidative stress and in wheat leaves they have been shown to be the main target of

oxidative stress (Bartoli et al., 2004). As targets of oxidative damage, processes within

plant mitochondria can be severely inhibited (Taylor et al., 2004). Key modulators of

the stress response such as salicylic acid and nitric oxide inhibit respiration in plants

39

(Millar et al., 2001). Furthermore various stresses are known to inhibit protein import

into mitochondria that in turn will affect several mitochondrial processes (Taylor et al.,

2003; Van Aken et al., 2009). Reactive oxygen species are most often a product of

stress. These products have been shown to cause oxidative damage to lipids,

proteins and nucleic acids (Gadjev et al., 2006). In mitochondria the main oxidative

damage targets appear to be Fe-S and lipoic acid containing enzymes, such as those

found in the respiratory chain components and the TCA cycle components

respectively (Taylor et al., 2004). Oxidative damage within the mitochondria can result

in the direct inhibition of Complex I and aconitase (Zhang et al., 1990; Verniquet et al.,

1991). The modification of lipoic acid residues has been shown to inhibit mitochondrial

pyruvate dehydrogenase (PDC) and 2-oxoglutarate dehydrogenase (OGDC) (Millar

and Leaver, 2000).

A possible reason why mitochondria are one of the main targets of oxidative damage

could be because they are capable of preventing the excessive build-up of ROS. If the

production of ROS was to be left unmonitored the likelihood of ROS induced cell

death is extremely high (Sun et al., 2002). There are known mechanisms within

mitochondria that suppress excessive ROS production. These include the Mn

superoxide dismutase (MnSOD) process that involves the conversion of superoxide

radicals to hydrogen peroxide and molecular oxygen. It has also been shown that the

resultant hydrogen peroxide can be further neutralised by mitochondrial ascorbate

peroxidase and gluthathione peroxidase (Chew et al., 2003; Navrot et al., 2006)

Another mechanism that prevents the excessive build-up of ROS includes the activity

of alternative oxidase and NAD(P)H dehydrogenases (NDs). Both these substances in

collaboration suppress ROS production by preventing the over-reduction of the ETC.

It has been found that a lack of AOX leads to a higher sensitivity to a combined stress

of drought and high light in Arabidopsis (Giraud et al., 2008) paired with an increase in

40

ROS formation (Vanlerberghe, 2013). These are clear demonstrations of the

importance of mitochondria in stress response and stress adaptation.

Mitochondria are not only a major target of oxidative stress they are also a major

source of cellular ROS. Complex I and Complex III of the electron transport chain are

involved in ubiquinone-ubiquinol inter-conversions, an intermediary of these reactions

is ubisemiquinone (Maxwell et al., 2002). It is thought that this is a major source of the

electrons that reduce molecular oxygen (O2) to O2- , H2O2 and the hydroxyl ionOH- is

the most damaging of all ROS species (Maxwell et al., 1999). ROS are in most cases

damaging to cells and toxic, however in some cases they can act as signals in stress

signalling cascades, second messengers during growth, movement and as a result of

hormone action (Suzuki et al., 2012). Thus, generation of ROS species in plant

mitochondria is an important mechanism by which the internal redox state can be

communicated to the nucleus and other cellular compartments.

1.10 Co-ordinated Expression of Nuclear and

Organellar Genes

The endosymbiotic theory on the origin of organelles as shown in Section 1.3

described the theory behind gene transfer. In Arabidopsis, only 33 proteins are

encoded in the mitochondria and only 128 proteins are encoded in the chloroplast,

and thus the majority of organellar proteins are encoded in the nucleus (Unseld et al.,

1997; Sato et al., 1999). In order for plants to function, a co-ordinated system between

the nucleus and organelles must occur. Therefore, nuclear encoded proteins are

transcribed in the nucleus, translated in the cytosol and then imported into their

corresponding organelle (McFadden, 1999; Van Aken et al., 2009). This multistep

pathway is controlled by anterograde mechanisms. In order for organelles to

communicate functional changes to the nucleus retrograde regulation is required. This

41

form of communication begins at the organelle and ends at the nucleus (Rhoads and

Subbaiah, 2007).

1.10.1 Anterograde regulation

Anterograde regulation has been described as “a forward flow” of information and has

also been described as a “top down” pathway of gene expression. There are many

questions regarding anterograde regulation and these include, “what are the

regulators of nuclear encoded organellar genes” and “how is this process regulated”.

From the complexity of these questions it is easy to understand that several levels of

anterograde regulation are required for the efficient functioning of these cells;

including the ability to change the level of transcripts of nuclear encoded mitochondrial

proteins (Kleffmann et al., 2004), control of transcript initiation, editing, processing and

maturation. Furthermore anterograde regulation controls almost entirely the

expression of the mitochondrial genome including DNA replication, transcription,

transcript processing and translation (Ng et al., 2014). The pentatricopeptide repeat

(PPR) family is the largest family encoding RNA-binding proteins in land plants and

they play an important role in anterograde regulation and gene expression. In

Arabidopsis there are approximately 450 family members and about 100 are predicted

to be targeted to organelle (Kotera et al., 2005). Other levels of anterograde regulation

include; the post translational import into the organelles, for chloroplasts the Tic and

Toc complexes are used and the Tim and Tom complexes are used for mitochondrial

import. This level of anterograde regulation is more complex for chloroplasts as the

import of proteins into the chloroplast is also dependent on the redox state of the

organelle, therefore further mechanisms that sense the redox state and respond to it

are involved (Küchler et al., 2002; Lister et al., 2004; Soll and Schleiff, 2004). Further

post translational actions require regulation from the nucleus and nuclear encoded

42

assembly factors, this often involves the assembly of multi protein complexes within

the mitochondria such as complex 1 (Shaughnessy et al., 2014).

1.10.2 Retrograde regulation

The mechanisms of retrograde regulation have evolved to communicate the functional

and developmental state of organelles to the nucleus, for this reason retrograde

regulation has been often referred to as the “bottom-up” pathway. The reason for this

form of communication may be as a result of alterations in organelle function from

multiple signals, stresses or mutations such as the presence of ROS or signalling

molecules or the availability of metabolites (Rhoads and Subbaiah, 2007). The

importance of this mechanism cannot be understated as it is crucial for resource

allocation under an altered organellar state (Woodson and Chory, 2008). An

interesting example of retrograde regulation can be seen in GUN mutants. The GUN1

mutant was mapped to a ~93 kilo base region of chromosome 2 and GUN1 alleles all

showed mutations in At2g31400 (Koussevitzky et al., 2007). Assays have shown that

GUN1 mutants were involved in tetrapyrrole biosynthesis (Kleine and Leister, 2016)

and the effects of Lincomycin on LHCB1.2 expression were suppressed in these

mutants (Gray et al., 2003). It is hypothesised that GUN1 mutants act in both

pathways and as a pentatricopeptide repeat-small MutS-related protein is presumably

involved in organelle gene expression (assuming that Norflurazon and Lincomycin

trigger different types of signalling) (Koussevitzky et al., 2007). Genetic evidence

suggests that GUN1 integrates signals derived from perturbations in the redox state,

chloroplast gene expression, and tetrapyrrole biosynthesis in seedlings. However, the

molecular mechanism by which GUN1 integrates retrograde signaling in the

chloroplast is unclear (Koussevitzky et al., 2007; Slonim and Yanai, 2009).

1.10.3 Mitochondrial Retrograde Regulation

43

Mitochondrial retrograde regulation (MRR) allows modulation of nuclear gene

expression in response to alterations in the mitochondrial state (Rhoads and

Subbaiah, 2007). This form of regulation is of critical importance for the smooth

functioning of organisms as mitochondrial dysfunction and the subsequent MRR is

thought to be a contributing factor in diseases such as Alzheimer’s, Parkinson’s,

Huntington’s, bipolar disorder, schizophrenia, Type II diabetes, cancer and the aging

process (Ben-Shachar and Laifenfeld, 2004; Butow and Avadhani, 2004; Beal, 2005;

Iwamoto et al., 2005; Kwong et al., 2006; Parihar and Brewer, 2007). MRR was first

demonstrated in yeast (Saccharomyces cerevisiae). The first target gene to be

identified of the retrograde pathway was shown in yeast to be CIT2, which encodes a

peroxisomal isoform of citrate synthase (Liu and Butow, 2006) and following genome

wide transcriptomic studies it has been shown that retrograde signalling is operational

in response to many signals in all eukaryotic organisms studied (Giraud et al., 2008;

Schwarzländer et al., 2011; Giraud et al., 2012; Quirós et al., 2016). AOX is the pre-

eminent model used to study MRR in plants (Rhoads and Subbaiah, 2007;

Vanlerberghe, 2013). AOX catalyses cyanide insensitive oxygen consumption and

oxidises its substrate without the generation of a proton motive force. At a

transcriptional, protein and activity level it is induced by a variety of abiotic and biotic

stresses, including photo-oxidative stress, temperature stress, nutrient deficiency,

water deficiency and oxygen deficiency (Vanlerberghe and McIntosh, 1997; Millar et

al., 2011; Ng et al., 2014). Several genes were shown to cluster with the expression

profile of AOX1a in response to perturbations tested in (Ng et al., 2014) including

Hsp60, sHSP23.5, sHSP23.6, MGE1, NDA2, NDB2 and UCP5, transporters and

several small auxin-up RNA (SAUR)-like proteins.

A prominent group of genes have been identified as all containing a cis-acting

regulatory element in their promoters to arbitrate the MRR mediated induction by

NAM, ATAF1/2 and CUC2 (NAC) transcription factors (De Clercq et al., 2013).

44

Several genes belonging to this group were clustered with AOX1a (Ng et al., 2014).

This group has been named the Mitochondrial Dysfunction Stimulon (MDS) and

UPOX1 is a member along with MGE1 (At5g55200), sHsp23.5 (At5g51440) and

AtOM66 (At3g50930) (Giraud et al., 2009; Van Aken et al., 2009; De Clercq et al.,

2013; Van Aken et al., 2013; Ng et al., 2014).

1.10.4 Organelle communications between mitochondria

and chloroplasts

Mitochondria and chloroplasts have an interdependent requirement on each other for

the survival of a eukaryotic cell. They are linked metabolically, because the

mitochondria oxidise the carbon that is fixed by the chloroplast during photosynthesis

and the chloroplast requires the ATP produced from mitochondrial dark respiration

(Leister, 2005). Although no known signalling molecules have been identified there

are several molecules implicated in playing a role in this organellar cross talk. ROS,

ascorbate and nitric oxide are all thought to play some role in the cross talk between

these organelles. In addition to these it is known that mitochondria produce redox

equivalents similar to that of the chloroplast, particularly the protectants against photo-

inhibition (Raghavendra and Padmasree, 2003; Lindemann et al., 2004). There are

also mutant lines that show indications of cross talk between these organelles. In

Arabidopsis AOX1a deficient mutants exhibited a high light and drought sensitive

phenotype with increased non-photochemical quenching and ROS levels. These

mitochondrial mutants also exhibited a decrease in transcript abundance for

photosynthetic and photo-respiratory transcripts. This is good evidence for organellar

cross talk, however, it still remains inconclusive, as these observations could have

been caused by indirect mechanisms (Giraud et al., 2008).

Recently, (Yoshida and Noguchi, 2011) have shown that under excess light reductant

are exported from the chloroplasts to the cytosol. It is hypothesised that this

45

communication occurs via NADP-Malate dehydrogenase (NADP-MDH)

and Malate-OAA shuttles and the mitochondrial non-phosphorylating pathways may

facilitate the dissipation of this excess reductant in the cell. Observations of organellar

communications were also observed in other species including maize, barley and

tobacco. The NCS6 maize mutant has a dysfunctional cytochrome oxidase subunit 2,

this consequently results in a dysfunction of the mitochondrial ETC and it has been

observed that this mitochondrial mutation corresponds with a decrease in

chloroplastic and nuclear encoded subunits of Photosystem I (Jiao et al., 2005). The

albostrians mutants in barley lack chloroplastic activity and it has been shown that this

corresponds with an increased mitochondrial gene copy number and an increase in

mitochondrial transcripts (Hedtke et al., 1999). Finally another mutant in tobacco, the

CMSII mutant that lacks the NAD7 subunit of complex I within the mitochondria has

been linked with a reduced rate of photosynthesis in dark to light transitions (Sabar et

al., 2000; Dutilleul et al., 2003).

1.11 Project Aims and Approaches

The best characterised processes in mitochondria are the production of ATP in the

process of oxidative phosphorylation and the TCA cycle. However mitochondria are

also critically important in plant defense in stress situations. Over the years many

mitochondrially located proteins have been identified as being induced by stress such

as AOX and NDB2, however the vast majority of mitochondrial proteins are yet to be

fully characterised.

This project aims to investigate the expression patterns and function of UPOX1 (Up

regulated due to OXidative stress). Prior to the start of this PhD, UPOX1 had not been

studied in depth. The first publications that used UPOX1 were studies by Clifton et al.,

2005, these experiments demonstrated that UPOX1 responded to a broad range of

abiotic stress treatments in cell cultures. Following this, the gene was named UPOX1

46

and described as one of the “hallmarks of oxidative stress” within the model plant

Arabidopsis thaliana (Gadjev et al., 2006). Apart from these transcriptomic studies no

in-depth transcriptomic or proteomic studies have been performed.

This project takes a multi-disciplinary approach to examining the induction of UPOX1

at a transcript and proteomic level with the eventual aim of elucidating the function of

UPOX1. The approaches taken include:

Transcript expression profiling-

This will involve investigations into the kinetics of UPOX1 induction with the aim of

identifying pathways that regulate stress induced gene expression. These assays will

be compared to that of known stress responsive genes such as AOX1a and other

mitochondrial proteins such as external NAD(P)H-dehydrogenase NDB2 and AtOM66.

In addition to this, transcript analysis will be conducted on defense signalling mutants

to further allude to the pathways that regulate stress induced gene expression.

Furthermore promoter regions will be investigated to determine the functional CAREs

that play a role in the response to stress treatments such as with H2O2, rotenone,

salicylic acid and combinations of these.

Proteomics -

Various proteomic methods will be employed including in vivo, in vitro localisation

methods and gel based isolation methods to further understand the proteomic profile

of UPOX1. Furthermore, transgenic lines that are over-expressers and under-

expressers for UPOX1 will be used to assist in determining the location of UPOX1

with the use of an antibody raised against UPOX1.

Global Transcriptomics –

In order to gain insights into the changes occurring within the transcriptome,

microarrays will be performed on the UPOX1 under-expressed and over-expressed

lines in comparison with Col-0 wild-type lines. Experiments will be conducted under

47

stress conditions and normal conditions. These microarrays may provide us with

more insight on the interactions and function of UPOX1 and associated transcripts.

48

49

50

Chapter 2. Materials and Methods

51

Chapter 2

MATERIALS AND METHODS

Chapter 2. Materials and Methods

52

2.1 General Chemicals

All reagents were molecular biology grade, analytical grade or equivalent, purchased

from Sigma-Aldrich (Sydney, Australia), Amersham Biosciences (Sydney, Australia)

Merck (Melbourne, Australia), Becton Dickinson (Melbourne, Australia), Bio-Rad

(Sydney, Australia), Univar APS Finechem (Melbourne, Australia) and AppliChem

BioChemica (Melbourne, Australia). Composition of solutions and media are listed in

Appendix 1. Water used for nucleic acid manipulations and water soluble solutions

was obtained from the Milli-Q Plus Ultrapure system (Millipore, Sydney, Australia) and

sterilized by autoclaving prior to use (denoted as SDW).

2.2 Arabidopsis Growth Conditions

2.2.1 Arabidopsis suspension cell culture

Arabidopsis thaliana suspension cell cultures (ecotype Landsburg erecta) (Sweetlove

et al., 2002) were cultivated in 100 mL cell culture media (Appendix 1) in 250 mL

flasks. Cell cultures were grown under long day light cycles of 16 h light

(~ 100 μE -2 s-1) and 8 h dark, rotating constantly at 120 revolutions per minute (rpm)

at 22°C. Cells were sub cultured at 7 day intervals with a 1:6 (v/v) dilution factor.

2.2.2 Arabidopsis water culture

Arabidopsis water cultures were cultivated in 80 mL water culture media (Appendix 1)

in 200 mL water culture containers. Cultures were grown under medium day light

cycles 12 h light (~ 100 μE -2 s-1) and 12 h dark, rotating constantly at 105 rpm at

22 °C.

2.2.3 Arabidopsis plant material

Arabidopsis plants were grown from seeds either in (4:1) soil mix : vermiculite or on

Arabidopsis solid media (Appendix 1) containing 50 µg/mL Cefotaxime® antifungal

Chapter 2. Materials and Methods

53

agent. Seeds were sterilized in a laminar flow hood using aseptic technique prior to

plating on solid media. Seeds were incubated in 1 mL sterilization solution (Appendix

1) for 5 min; this solution was removed and incubated in 1 mL 70% (v/v) ethanol. The

70% ethanol solution was removed and the seeds were washed in 100% ethanol

before being dried in the laminar flow. Seeds were resuspended in 200 μL of sterile

distilled water (SDW) and transferred to solid media. To synchronize germination,

seeds plated on solid media and in soil mix were stratified at 4 °C in the dark for 48 h,

and then grown at 22°C under long day conditions of 16 h light (~ 100 μE -2 s-1) and 8

h dark.

2.2.4 Chemical treatment of Arabidopsis cell culture,

water culture and plant material

Arabidopsis cell cultures were treated four days after subculturing. The following table

shows how the treatments were prepared and applied. The following treatments were

applied according to (Clifton et al., 2005).

Treatment Solvent Final

Concentration

Quantity Applied

Antimycin A ethanol 5 μM 12μLof 50μM

Glucose H2O 3 % (w/v) 1 mL of 2.17 M

H2O2 30% (v/v) 10 mM 130 μL of 8.82 M

Rotenone ethanol 40 μM 480 μL of 10 mM

Salicylic acid ethanol 100 μM 120 μL of 100 mM

All compounds were obtained from Sigma (Castle Hill, Australia).

2.2.5 UV treatment of Arabidopsis water culture

Chapter 2. Materials and Methods

54

Arabidopsis water cultures were treated with UV stress by exposing them to UV-C at

254 nm using cross linker (UVP 34-0042-01). Following irradiation, plants were

incubated under the same conditions used for routine growth unless stated otherwise

2.3 Bacterial Techniques

2.3.1 Bacterial cell strains

JM109:

recA1supE44endA1hsdR17gyrA96relA1thi(lac-proAB)F’[traD36proAB+,lacIqlacZM

15]

DH5:

supE44lacU169(80dlacZM15)hsdR17recA1endA1gyrA96thi-1relA1

XL1-blue:

supE44hsdR17recA1endgyrA46thirelA1lacF’[proAB+lacIqlacZ∆M15Tn10(tert)

XL10-Gold

Tetr∆(mrcA)183∆(mcrCB-hsdSMR-mrr)173endA1supE44thi-

1recA1gyrA96relA1lacHte[F’proAB+,lacIqZ∆M15Tn10(tetr)AmyCamr]

2.3.2 Transformation into JM109 and DH5 cells

Chemically competent cells (laboratory competent JM109 or DH5) were thawed on

ice, mixed gently and 50 μL were transferred to pre-chilled 15 mL round bottom tubes.

Up to 5 μL of DNA, containing up to 50 ng of plasmid DNA was added to the cells.

The transformations were incubated on ice for 10 min, heat shocked for 45 sec in a

water bath at 42°C and incubated on ice for a further 2 min. 500μL of LB media (for

JM109 cells) or 200 μL SOC media (Appendix 1) (for DH5) were added to the cells

and the transformations were incubated at 37°C for 1 hr, shaking at 220 rpm. The

transformations were then divided into 50 μL and 450 μL; these volumes were plated

Chapter 2. Materials and Methods

55

onto LB plates with the appropriate antibiotics (Kanamycin or Ampicillin) and

incubated overnight at 37°C.

2.3.3 Transformation into XL-Blue Super competent cells

Prior to transformation 4 units (U) of Dpn1 restriction enzyme was applied to the

polymerase chain reaction (PCR) reaction. 200 ng of the Dpn1 digested DNA was

added to 25μL of XL- Blue super competent cells and incubated for 30 min on ice in a

pre-chilled 15 mL round bottom tube. Cells were then heat shocked for 45 s at 42 °C

and incubated on ice for 2 min. Five hundred μL of pre-warmed NZY+ broth was

added to the transformations and they were incubated for 1 h at 37°C, shaking at 220

rpm. 50μL and 450 μL were plated onto separate LB plates with the appropriate

antibiotics (Kanamycin or Ampicillin) and incubated overnight at 37 °C.

2.3.4 Transformation into XL-Gold Ultra competent cells

Chemically competent cells were thawed on ice and 25 μL of cells were transferred to

pre-chilled 15 mL round bottom tubes. One μL of 50% (v/v) β-mercaptoethanol, were

added to the cells and incubated for 10 min on ice. Two to five μL of Dpn1 digested

DNA was added to the cells and mixed gently. The cells were heat shocked for 30 s at

42°C and incubated on ice for a further 2 min. 500μL of pre-warmed NZY+ broth was

added to the transformations and they were incubated for 1 h at 37°C, shaking at 220

rpm. Fifty μL and 450 μL were plated onto separate LB plates with the appropriate

antibiotics (Kanamycin or Ampicillin) and incubated overnight at 37 °C.

2.4 Nucleic Acid Manipulation

2.4.1 Genomic DNA preparation

Genomic DNA was isolated from Arabidopsis tissue using DNAeasy® Plant Mini Kit

(QIAGEN, Clifton Hill, Australia) as per manufacturer’s instructions.Plant samples

Chapter 2. Materials and Methods

56

were mechanically disrupted and chemically lysed, during the lysis step RNA was

removed by RNase digestion. Following the removal of cell debris the samples were

filtered by centrifugation through QIAshredder spin columns. Binding buffers and

ethanol were washed through the spin columns to promote the binding of DNA to the

membrane and remove contaminants. The precipitating proteins, polysaccharides and

lysate were then centrifuged through further spins. The DNA selectively bound to the

silica-gel membrane and contaminants passed through. Two further wash steps

ensured highly purified DNA. The DNA was finally eluted in 100 μL of SDW and

stored at -20°C.

2.4.2 Total RNA isolation

Total RNA was isolated using the RNeasy Plant Mini Kit (QIAGEN, Clifton Hill,

Australia). For each sample, approximately 100 mg of Arabidopsis plant tissue was

ground under liquid nitrogen with a mortar and pestle. These samples were then lysed

and homogenized in a highly denaturing guanidine-thiocyanate–containing buffer, this

inactivates RNases. Ethanol was added to the samples to promote the binding of RNA

to the RNease mini spin columns. The RNA bound to the membranes selectively

whilst contaminants were washed through. All reactions were treated with RNase-free

DNaseI (QIAGEN, Clifton Hill, Australia) and DNAfree™ (Ambion, Austin, USA)

according to manufacturer’s instructions to ensure the complete removal of any DNA

contamination. Ethanol precipitations were performed for all RNA samples extracted

from MS plates due to the high sugar content. RNA samples were eluted in80 μL of

RNase-free SDW and further diluted with 120μL of SDW. 20 μL of sodium acetate

was added with 440 μL of 100% (v/v) ethanol to the RNA. This reaction was kept at -

20°C for at least 2 h and centrifuged at 20 000 g for 25 min at 4°C. The supernatant

was removed and the remaining pellet was washed in 1 mL of 70% (v/v) ethanol,

centrifuged at 20 000 g for 20 min at 4°C and dried in a speedy-vac for 10 min. The

Chapter 2. Materials and Methods

57

pellet was resuspended in 40 μL of RNA-free SDW. RNA intended for qRT-PCR and

microarrays required stringent quality and quantity assessment. This was assessed

on 1.2 % (w/v) agarose gels and was based on the integrity of the 18S and 28S rRNA

bands. Quantity checks were also analysed using a Nanodrop ND-1000

Spectrophotometer (Biolab, Victoria, Australia) and further quality analysis was

performed using a Bioanalyzer 1200 (Agilent, California, USA).The A260/A280ratio

obtained using the Nanodrop and the Bioanalyser gave an indication of the purity of

the nucleic acid, typically ratios between 1.8 and 2.1 were considered pure.

2.4.3 Preparation of cDNA

Reverse transcription was performed to produce cDNA from total RNA. Using the

iScript cDNA synthesis kit (Bio-Rad, Sydney, Australia) 1 μg of total RNA was used

and cDNA was produced according to manufacturer’s instructions. To produce cDNA

an enzyme was used that contained a modified MMLV-reverse transcriptase with

RNase H+ activity. Also with the addition of SDW a five times transcription mix was

added to the reactions that contained RNase inhibitor, a blend of oligo(dT), random

hexamer primers and dNTPs. The reactions were incubated at 25 °C for 5 min, 42 °C

for 30 min, 85 °C for 5 min and held at 4 °C.

For qRT-PCR assays a negative control was always performed, these reactions

contained all the components described above except the reverse transcriptase

enzyme.

The cDNA samples were purified prior to quantitative PCR analyses, using the

QIAquick® PCR Purification Kit (Qiagen, Clifton Hill, Australia) as per manufacturer’s

instructions. A binding buffer was applied to the cDNA samples, this is a high-salt

buffer, DNA binds selectively to the silica membrane within the spin column. The wash

buffers wash all primers, nucleotides, enzymes and other impurities through the

column leaving the bound DNA samples attached to the membrane. Finally the cDNA

Chapter 2. Materials and Methods

58

samples were eluted in 30 µL of low salt buffer and stored at -20 °C. All samples to

be used in qRT-PCR were then diluted 1:10 in SDW.

2.4.4 Agarose gel electrophoresis

Agarose gels electrophoresis was used to analyse PCR products and restriction

digests by DNA separation. Nucleic acids were separated using 1% (w/v) agarose

(Roche, Castle Hill, Australia) and gels were made in 1X TAE buffer (Appendix 1)

containing 1 µg/mL ethidium bromide. After the Agarose gels solidified the samples

were mixed with loading buffer (Appendix 1) and electrophoresis took place in tanks

containing 1X TAE buffer under 70 Volts (V). Following electrophoresis, samples were

visualised under UV light using a gel doc system (Bio-Rad, Sydney, Australia).

2.4.5 Amplification of DNA by polymerase chain reaction

The Expand High Fidelity Polymerase Chain Reaction kit along with the dNTP kit

(Roche, Castle Hill, Australia) was used under manufacturer’s instructions to perform

all polymerase chain reactions (PCR). Primers were diluted to 20 ρmol/μL (Sigma-

Aldrich, Sydney, Australia; Invitrogen, Sydney, Australia) prior to PCR. Amplifications

were performed under the following parameters: 94°C for 2 min followed by 35 cycles

of 94°C for 30 s, 58°C for 2 min and 72°C for 5 min. Small adjustments were made to

the annealing temperature depending on the melting points of individual primers.

Following PCR, reactions were held at 4°C prior to purification. PCR products were

purified using the QIAquick PCR purification kit (QIAGEN, Clifton Hill, Australia) as

described in Section 2.4.3.

2.4.6 TOPO-TA cloning into pCR2.1

Following PCR purification TOPO-TA cloning kits (Invitrogen, Sydney, Australia) were

used to clone PCR products into the pCR2.1 vectors according to manufacturer’s

instructions. 4μL of PCR product was incubated with 1 μL of salt solution (consisting

Chapter 2. Materials and Methods

59

of 1.2 M NaCl and 0.06 M MgCl2) and 1μLof 10 ng/μL of vector for 10 min at room

temperature. 2μL of this reaction was transformed into the TOP10F’ cells supplied

with the TOPO-TA kit. The cells were then heat shocked for 30 s at 42°C and

incubated for 2 min on ice. 250μL of SOC media was added to the transformations

and incubated for 1 h at 37°C, shaking at 220 rpm. 40μL of 40 mg/mL X-gal (5-bromo-

4-chloro-3-indolyl-β-D-galactopyranoside) and 40 μL of 40 mg/mL IPTG (isopropyl-1-

thio-β-D-galactopyranoside) were added to LB plates containing the appropriate

antibiotics (Kanamycin or Ampicillin) prior to the transformations being plated.

2.4.7 Site Directed mutagenesis

Site directed mutagenesis (SDM) was performed using the QuickChange II or the

QuickChange XL II site directed mutagenesis kits (Stratagene, California, USA).

Generally QuickChange XL II kits were used for mutations that included more than 3

point mutations or for reactions that had been previously unsuccessful using the

QuickChange II kit. Mutagenesis was then performed according to the manufacturer’s

instructions as follows. Mutagenesis reactions used supercoiled double stranded DNA

(dsDNA) vectors, primers, dNTP mix, a 10 times reaction buffer, SDW and PfuTurbo

DNA polymerase. Primers were purchased from Invitrogen (Sydney, Australia) and

were designed with the desired mutation in both the forward and reverse orientations.

Cycling parameters were as follows: 95 °C for 1 min followed by 18 cycles of 95 °C

for 50 s, 55 °C for 50 s, 68 °C for 1 min/kb of plasmid followed by 68 °C for 7 min.

Cycling parameters were similar between with QuickChange II and the QuickChange

XL II, the major difference was the QuickChange II kit did not contain the final cycle of

68 °C for 7 min, all reactions were held at 4 °C.

2.4.8 Dpn1 Digestion

Chapter 2. Materials and Methods

60

Mutagenesis reactions were digested using 4U of Dpn1 endonuclease and incubated

at 37 °C for 1.5 hr. Dpn1 endonuclease removes parental, unmutated strands by

specifically digesting methylated and hemi-methylated DNA leaving only daughter

DNA containing the desired mutation. Following Dpn1 digestion SDM reactions were

transformed using XL-Blue Super competent cells (for QuickChange II reactions) or

XL-Gold Ultra competent cells for (QuickChange XL II reactions) according to Section

2.3.3 and Section 2.3.4.

2.4.9 Restriction digests

Restriction digests for sub-cloning purposes required ~1000 ng of purified insert DNA

and ~1000 ng of vector DNA. Digestions were set up using the appropriate restriction

buffers and enzymes depending on what the restriction site(s) were (Invitrogen,

Sydney, Australia). The digest reactions were made up to 30 μL and incubated at the

appropriate temperatures (most commonly 37 °C) for 2 hr. The restriction digests

were then incubated at 65 °C for 15 min to denature the enzymes. Following

inactivation of enzymes, the insert DNA along with 1 μL of vector DNA were analysed

using 1% (w/v) agarose gel electrophoresis using a DNA ladder containing known size

fragments (Invitrogen, Sydney, Australia).

2.4.10 Calf Intestinal Alkaline Phosphatase treatment

For sub-cloning, vector digests were treated with Calf Intestinal Alkaline Phosphatase

(CIAP) (Invitrogen, Sydney, Australia). CIAP prevents re-ligation of linearized vector

DNA by removing 5'-phosphate groups prior to 5'-end labelling of nucleic acids. Blunt

ended vector DNA digests were incubated at 37 °C for 1 hr, 5’ overhang vector

digests were incubated at 37 °C for 30 min. Following CIAP treatment any remaining

CIAP enzyme was inactivated at 65 °C for 15 min.

Chapter 2. Materials and Methods

61

2.4.11 DNA ligation

Following the PCR purification of the insert DNA and the CIAP treatment of the vector

DNA both samples were analysed using a Nanodrop to determine their DNA

concentrations. Calculations were performed to in order to obtain a 3:1 insert to vector

molar ratio in the ligation reaction. The formula for working out the correct molar ratio

is as follows: ((ng vector) x (kb size of insert)) / ((kb size of vector) x (molar ratio of

(insert/vector)) = (ng insert). T4 ligase buffer was added to a final concentration of 1 x

and 1 U of T4 ligase was added (Roche, Castle Hill, Australia). The reactionswere

incubated at 14 °C overnight prior to transformation with chemically competent cells.

2.4.12 Plasmid Minipreps

Single E.coli bacterial colonies containing a plasmid were used to inoculate 2 mL of

LB media containing appropriate antibiotics. The cultures were grown for 16 h at

37 °C with shaking at 1400 rpm. Cells were centrifuged for 1min at 20 800 g, the

supernatant was removed and the plasmid DNA was isolated from the cell pellet using

the QIAGEN Plasmid Mini kit (QIAGEN, Clifton Hill, Australia) according to

manufacturer’s instructions. The process by which QIAGEN Plasmid Mini kits purify

DNA involved the use of spin columns made with anion-exchange resin. Following

the lysis of the bacterial pellets the alkaline lysate was applied onto the anion-

exchange resin tip, under centrifugation and low salt conditions the lysis was cleared

leaving the DNA bound to the membrane. RNA proteins, dyes, and low-molecular-

weight impurities were then removed by the addition of a medium-salt wash under

centrifugation and plasmid DNA was eluted in a high salt buffer. The DNA was

desalted and concentrated following isopropanol precipitation and collected by

centrifugation to be finally eluted in 20 μL of SDW and stored at -20 °C.

2.4.13 Plasmid Midipreps

Chapter 2. Materials and Methods

62

Single E.coli bacterial colonies containing a plasmid were used to inoculate 50 mL of

LB media containing appropriate antibiotics. The cultures were grown for 16 h at

37 °C with shaking at 220 rpm. Cells were collected by centrifugation for 15 min at

3000 g. The plasmids were then purified using the QIAGEN Plasmid Midi kit

(QIAGEN, Clifton Hill, Australia) as per manufacturer’s instructions. The principles by

which the QIAGEN Plasmid Midi kit are based on involve the use of a anion-exchange

resin column and selective binding under varying salt and pH conditions that operate

under gravity flow, they are the same principles as that described for QIAGEN

Plasmid Mini kit and the process occurs as described in Section 2.4.13. The key

differences involve the use of larger liquid quantities for the required binding, washing

and elution buffers. The DNA was eluted in 100 μL of SDW and stored at -20 °C.

2.4.14 Plasmid Maxipreps

Single E.coli bacterial colonies containing a plasmid were used to inoculate 150 mL of

LB media containing appropriate antibiotics. The cultures were grown for 16 h at 37

°C with shaking at 220 rpm. Cells were collected by centrifugation for 30 min at

3000 g. The plasmids were then purified using the QIAGEN Plasmid Maxi kit

(QIAGEN, Clifton Hill, Australia) as per manufacturer’s instructions. The principles by

which the QIAGEN Plasmid Maxi kit are based on involve the use of a anion-

exchange resin column and selective binding under varying salt and pH conditions

that operate under gravity flow, they are the same principles as that described for

QIAGEN Plasmid Mini and Midi kits and the process occurs as described in Section

2.4.13. The key differences involve the use of larger liquid quantities for the required

binding, washing and elution buffers. The DNA was eluted in 1 ml of SDW and stored

at -20 °C.

2.5 Generation of Arabidopsis mutant lines

Chapter 2. Materials and Methods

63

2.5.1 Preparation of Artificial microRNA constructs

For Arabidopsis stable transformations the experimental procedure was carried out

according to the Web MicroRNA Designer 2.0 (Schwab et al., 2006). The artificial

microRNAs (AmiRNAs) were generated by overlapping PCRs and the MIR319a

precursor was used as the plasmid to engineer the AmiRNA. The AmiRNA fragment

was sub-cloned from the MIR319a precursor into the pRS300 plasmid and

consequently sub-cloned into the pCambia1301 plasmid for agrobacterium mediated

transformation according to Section 2.4.9 to Section 2.4.12.

2.5.2 Preparation of Agrobacterium tumefaciens electro-

competent cells

Two hundred μL of Agrobacterium stock was inoculated with 10 mL LB and

Rifampicin to a final concentration of 50 μg/mL. This culture was grown over night at

30 °C shaking at 220 rpm. One hundred μL of overnight culture was added to 900 μL

of LB with Rifampicin and the amount of starting culture required to inoculate 100 mL

of LB with Rifampicin was calculated. The 100 mL culture was grown over night at

30 °C shaking at 220 rpm until the optical density was between 0.4 and 0.6. This

culture was centrifuged at 4 °C for 10 min at 10 000 rpm and the pellet was

resuspended in 10mLice cold1 mM HEPES buffer. The centrifugation was repeated

and the pellet was resuspended in 10mLice cold of 1 mM HEPES, 10 % (w/v)

glycerol. The centrifugation was repeated, the resultant pellet was resuspended in

10mL of 1 mM HEPES 10 % (w/v) glycerol and the suspensions were pooled. The

solutions were centrifuged at 4°C for 5 min at 12 200 g and the pellet was

resuspended in 200 μL of 1 mM HEPES, 10 % glycerol. The samples were aliquoted

into 100 μL samples and snap frozen at -80°C.

2.5.3 Transformation of electro-competent

Agrobacterium

Chapter 2. Materials and Methods

64

One hundred μL of Agrobacterium competent cells were thawed on ice and 2 μL of

plasmid (200-400ng) was added, the reaction was chilled on ice for 2 min. Then

transferred to a cuvette and 500 μL of LB was pipetted and immediately pulsed using

the Gene Pulser (Bio-Rad, Sydney, Australia). The cells were shocked with 2.5 kV,

with a capacitance of 25 μF and a resistance of 400 Ω. Immediately following

electroporation, 500 μL of LB was added and the reaction was transferred to a 15 mL

round bottom tube and incubated for 2 h at 30 °C shaking at 220 rmp. 50 μL and

200 μL of culture were plated onto LB plates containing Rifampicin in conjunction with

other appropriate antibiotics (Kanamycin or Ampicillin).

2.5.4 Floral dipping

Based on the methods detailed in (Clough and Bent, 1998) Arabidopsis plants were

grown until they started to bolt and seven days prior to transformation the bolts were

clipped. The agrobacterium was inoculated in 5 mL of LB with required antibiotics and

grown for 2 days at 28 °C shaking at 200 rpm. The small culture was transferred to

500 mL of LB media with required antibiotics and grown overnight at 28 °C, shaking at

200 rpm until the optical density reached 0.6 ~ 0.8. The agrobacterium was then

centrifuged for 20 min at 2500 g and resuspended in 500 mL of 5 % (w/v) sucrose,

0.03% (v/v) silwet L-77. The above soil parts of the Arabidopsis plants were dipped in

the agrobacterium for 10 s with gentle agitation. The remaining solution was pipetted

onto the leaves. The plants were covered for one to two nights to maintain high

humidity levels. The plants were grown as usual and the dry seeds were harvested

and the transformants were selected using hygromycin selection on MS agar plates.

2.5.5 Preparation of mutant lines

The full length coding sequence of UPOX1 was cloned into the p35S overexpression

vector pK7WG2 and homozygous transgenic plant lines were created by Dr Van Aken

Chapter 2. Materials and Methods

65

and Dr Inge De Clercq within Prof. Frank Van Breusegem's lab (VIB Ugent, Belgium),

this line was provided for this collaboration. This line was also confirmed for

homozygosity within the Whelan lab by myself prior to qRT-PCR analysis.

Promoter:GFP-GUS lines were generated by isolating DNA from Col-0 plants with

DNeasy plant kits (Qiagen) according to the manufacturer's instructions. Following

amplification of the 1500 bp genomic region upstream of the start codon with Platinum

Taq High Fidelity DNA polymerase (Invitrogen), the PCR product was cloned into

pDONR221 (Thermofisher, Melbourne Australia) and cloned by recombination to

pBGWFS7 (VIB-Ghent University, Ghent, Belgium) (Karimi et al., 2002), generating a

transcriptional GFP-GUS fusion. These constructs were then transformed into

Arabidopsis Col-0 by Agrobacterium tumefaciens-mediated floral dipping.

2.6 Transient transformation of Arabidopsis

cell culture and leaves

2.6.1 Preparation of Arabidopsis suspension cells

Arabidopsis cell culture was harvested at 4 days and stress treatments were applied

in the manner described in Section 2.2.4 for 1 hr. Cells were aseptically syringed onto

filter paper and left for 2 hours on solid osmotic media (Appendix 1) prior to biolistic

transformation.

2.6.2 Preparation of Arabidopsis leaves

Four week old rosette leaves were excised and stress treatments were applied in the

manner described in Section 2.2.4 for 3 hr. Leaves were placed on filter papers which

were then placed on solid osmotic media prior to biolistic transformation.

2.6.3 DNA precipitation onto gold microcarriers

Chapter 2. Materials and Methods

66

To a 1.5 ml tube 30 mg of 1 µm gold particles was aliquoted, this was washed in 1 ml

of 70% ethanol and following the removal of the ethanol the gold particles were

resuspended in 500 µl of 50% glycerol. The gold particles were aliquoted into 50 µl

lots. To tube, 5 µl of DNA (at 1 µg/µl), 50 µl 2.5 M CaCl2 and 20 µl of 100 mM

Spermidine was added prior to biolistic transformation. After vortexing and

precipitation, the gold beads were washed with ethanol.

2.6.4 Transformation of tissue

Using the PDS-1000 (Bio-Rad, Sydney, Australia) transformations of each construct

were performed in triplicate according to manufacturer’s instructions. A hepta adapter

was used and all transformations were performed under a vacuum pressure of 27

inches/Hg and a helium pressure of 1300 psi. The rupture disks (Bio-Rad, Sydney,

Australia) were rated at 1100 psi. Following transformation of cell culture and leaves

they were placed under light at 22 °C for 24 hr.

2.6.5 Reporter gene assays for luciferase and

β-glucuronidase

Tissue was ground under liquid nitrogen using stainless steel ball bearings and the

Mixer Mill 301 (Retsch, Haan, Germany). The tissue was extracted according to the

manufacturer’s instructions using the lysis buffer supplied with the Luciferase Assay

System (Roche, Sydney, Australia).The Polarstar Optima (BMG Lab technologies,

Offenburg, Germany) was used to perform Luciferase (LUC) and 4-methylumbelliferyl

β-D-glucuronide dehydrate (MUG) activity assays. Activities for LUC were measured

at 4 s after the addition of substrate. MUG activities were assayed by measuring the

accumulation of MUG fluorescent product over 1 h with measurements at 1 min

intervals using a fluorimetric assay described by (Jefferson et al., 1987). All samples

were excited at 355 nm and emission was measured at 460 nm.

Chapter 2. Materials and Methods

67

2.6.6 Normalisation of LUC and MUG activities

An estimation of the transformation efficiency was provided by dividing the LUC

activities by the highest LUC value for the set. All MUG values were consequently

divided by this value to give a normalized MUG value. Standard errors were then

calculated by dividing the standard deviation by the square root of the number of

transformations performed for each construct.

2.7 Quantitative RT-PCR

2.7.1 DNA standards

Gene specific DNA standards are required in order to enable the measurement of

absolute cDNA abundance for quantitative real time polymerase chain reaction

(qRT-PCR). The gene of interest was cloned using cDNA as a template and specific

forward and reverse cloning primers. The DNA standards were then amplified via

PCR. The PCR products were separated by agarose gel electrophoresis and purified.

The amount of DNA was quantitated using the PicoGreen® dsDNA quantitation kit

(Molecular Probes, Eugene, USA) according to manufacturer’s instructions. DNA

standards were kept at 0.1 fmol/μL and diluted to 0.01 fmol/μL as a working stock.

2.7.2 Primer design and optimization

Primers for qRT-PCR were all designed to be intron spanning with an amplicon size of

approximately 200 bp in size. Gene specificity tests were conducted to ensure no

cross amplification for genes within a family, the primers used for qRT-PCR are listed

in Appendix 2.

The maximum efficiency for the qRT-PCR reactions were individually tested by using

serial dilutions of the standard DNA template, (by varying the Mg2+ concentration and

the primer concentrations). Following qRT-PCR runs on the Light Cycler® 480 (Roche,

Chapter 2. Materials and Methods

68

Castle Hill, Australia) melt curve analysis was performed to determine if any primer-

dimer and/or non-specific amplification had occurred.

The absence of genomic DNA contamination was confirmed by assaying all no

reverse transcriptase reactions (no-RT). If any no-RT samples showed specific

amplification all samples were discarded and the process was repeated from the

beginning with new RNA.

2.7.3 qRT-PCR using the Roche Light Cycler® 480

Serial dilutions were prepared from the DNA standards, these ranged from

0.01 fmol/μL to 0.00000001 fmol/μL. Five μL reactions were assayed with varying

primer concentrations from 0.3 to 0.9 μM, to these reactions 2.5 μL of Roche SYBR

Green and 1 ul of cDNA was added. Prior to the commencement of all qRT-PCR runs

all plates were mixed gently and spun down at 1000 g for 1 minute.

More specific primer hybridization was provided by the pre-incubation step performed

at 95 °C for 10 min. The remainder of the qRT-PCR assayed consisted of the

following parameters: amplification for 45 cycles; 95 °C for 10 s, 60 °C for 10 s and

72°C for 10 s with a single data acquisition point. Melt curve consisted of the following

parameters: 95 °C for 10 s, 65 °C for 1.01 min, 95 °C continuous with 5 data

acquisitions per cycle and cooling at 40 °C for 10 s. The results were analysed using

the software on the Light Cycler® 480.

2.7.4 qRT-PCR Analysis

Prior to analysing the qRT-PCR results melt curve analysis was performed, the

principle behind melt curve analysis involves the denaturation of base-base hydrogen

bonds. The energy required for this break is dependent on the length, GC content and

Chapter 2. Materials and Methods

69

complementarity of a sequence. Therefore any non-specific products or primer

dimmers amplified during a qRT-PCR run can be detected by SYBR green.

The data analysis software calculated a threshold cycle for each sample, this

represents the cycle at which the fluorescence signal in the sample first increased

significantly above the background level. From the threshold cycles (Ct) of the

standard DNA dilution series a standard curve was generated between 0.01 fmol/μL

and 0.00000001 fmol/μL. This was then used to determine the starting concentration

of transcript in each cDNA sample based on its threshold cycle.

2.7.5 qRT-PCR Cross reactivity assays

Performed on the Roche Light Cycler, melt peaks confirmed if the primers were

specific for the target transcript. This tool plots temperature against fluorescence

units. The fluorescence increases as the temperature increases until a point at which

the dsDNA denatures to form ssDNA which involves unbinding the SYBR® Green dye

and losing fluorescence. This data is represented by plotting temperature against the

negative derivative of the relative fluorescence units. A single peak at a specific

temperature represents the specific product of uniform size. Any non-specific product,

such as primer dimmer could be visualized as a substantially smaller peak occurring

at a lower temperature. This method allowed products of different size to be

differentiated with 1 °C accuracy or 1 bp accuracy.

2.8 Organelle and protein isolation methods

2.8.1 Mitochondrial Isolation

All mitochondrial isolation steps were carried out at 4 °C and centrifugation was

performed with the use of the Beckman J-26XD and J301 centrifuges with JA-20

Chapter 2. Materials and Methods

70

rotors. All media, centrifuge tubes and apparatus were pre-chilled at 4 °C prior to use.

The procedure for mitochondrial isolation was based on the methods described by

(Day et al., 1985) as follows.

Mitochondrial isolations from water cultures and whole tissues involved the same

buffers and centrifugations spins, for isolation of mitochondria from cell cultures there

were slight variations in the buffers used and the centrifugation speeds, the principles

were however the same. Four week old plants grown on soil and 10-14 day old water

cultures were used for all mitochondrial isolations. Grinding and wash buffers were

made (Appendix 1) in advance and were kept at 4°C. On the morning of the

mitochondrial isolation 1.06 g (per 300ml media) of sodium ascorbate and 0.74 g (per

300ml media) of L-cysteine were added to the grinding media to stabilize the

mitochondria during the grinding process. The plant material was homogenized by

hand using mortar and pestles and filtered through 4 layers of miracloth, this process

was repeated twice. The filtered homogenate was centrifuged under 2 450 g for 5 min

to remove the cell debris and nuclei. The supernatant was transferred to new tubes

containing 1 times wash buffer and centrifuged at 17 400 g for 20 min to pellet the

mitochondria and peroxisomes. The pellets were resuspended in ~2 mL of 1 times

wash buffer and the previous low and high speed centrifugation steps were repeated

to further purify the crude mitochondrial fraction. Following this, the crude

mitochondrial pellets were layered over 0-40% (w/v) PVP gradients (Appendix 1).

These gradients were centrifuged for 40 min at 40 000 g. The PVP served to narrow

the distribution of mitochondria within the gradients, mitochondrial bands visibly form a

yellow/grey band low in the tube. The layers above were aspirated off and the

mitochondrial bands were distributed in 1 times wash buffer to remove the percoll and

other impurities. Following two high speed spins at 31 000 g for 15 min the final

mitochondrial pellet was isolated.

Chapter 2. Materials and Methods

71

Mitochondria following this isolation method was sufficiently pure to perform in vitro

imports however if mitochondria required further purification for downstream

application such as proteinase K titrations a second gradient was performed prior the

final high speed spins at 31 000 g.

2.8.2 Chloroplast Isolation

All pea leaf chloroplast isolation steps were carried out at 4 °C and centrifugation was

performed with the use of the Beckman J-26XD and J-301 centrifuges with JA-20

rotors. All media, centrifuge tubes and apparatus were pre-chilled at 4 °C prior to use.

The procedure for chloroplast isolation was based on (Bruce et al., 1994) as follows.

Prior to a chloroplast isolation the day/light cycle at 26 °C was set so as chloroplasts

were isolated at the end of a dark period, this prevents the accumulation of starch.

The peas harvested ranged in age between 7 and 10 days and the percoll gradients

(Appendix 1) were prepared in advance. The plant tissue was homogenized in two

times grinding buffer using a standard blender with 5-6 short bursts. The

homogenized material was then filtered through one layer of miracloth and centrifuged

at 2000 g for 5 min to pellet the crude chloroplasts. The resuspended pellet was then

pipetted over two gradients and centrifuged at 12 100 g. Chloroplasts form a light

green band low in the tube, this fraction was then washed and centrifuged at 2000 g

for 4 min. The resultant pellet was then assayed for its chlorophyll concentration prior

to use in in vitro imports.

2.8.3 Protein translation

Radiolabelled [35S]-methionine precursor proteins were translated using the TNT®

Coupled Transcription Translation Rabbit Reticulocyte system by Dr Murcha. As this

translation was weak it was enhanced by myself with two additional methionines at

the start and end of the protein (Stratagene, California, USA). This in vitro synthesis

Chapter 2. Materials and Methods

72

system provided a master mix containing reticulocyte lysate, RNA polymerase, amino

acids, buffer, RNase inhibitor and [35S]-methionine. Translations were all prepared on

ice, mixed with plasmid DNA, incubated for 2 h at 30 °C and frozen until use. Prior to

the use of translation in imports the translations were tested by running 2 μL on SDS-

PAGE gels (Section 2.9.7 SDS-PAGE) and exposing them on Fuji BAS TR2040

phosphor imaging plates.

2.9 In vivo and in vitro assays

2.9.1 In vivo GFP Assays

The GFP is composed of 238 amino acids, with a predicted mol mass of 26.9 kDa. It

absorbs blue light maximally at 395 nm with a minor peak at 470 nm and emits a

green light at 509 nm with a shoulder at 540 nm (Phillips, 2001).

The process involved in creating GFP fusions is relatively straight forward; this was

carried out via PCR and cloning strategies following the initial sub-cloning of UPOX1

into the pCR2.1 by Prof Ryan Lister (within the Whelan lab). The GFP fusions were

then subsequently sub-cloned by using the coding sequences (CDS) which were

amplified from Arabidopsis cDNA and cloning them into GFP fusion vectors, for

UPOX1, UPOX2 and UPOX3 both C-terminal (CDS-GFP) and N-terminal (GFP-CDS)

GFP fusions were constructed. The choice of the carboxy or amino terminals is

usually based on predicting which region of the proteins will tolerate the addition of

GFP and remain functional (Pédelacq et al., 2005). As this was a preliminary

examination of these proteins both terminals were used in individual constructs.

These fusion vectors were always co-transformed with RFP control constructs in order

Chapter 2. Materials and Methods

73

to define the sub cellular localisation of GFP with certainty. The RFP fusions used as

mitochondrial controls were AOX transit peptide red fluorescent proteins (AOX-RFP)

which were created by Dr Murcha and Dr Chris Carrie (Murcha et al., 2007; Carrie et

al., 2009).

These GFP and RFP fusions were transformed into Arabidopsis cell culture and onion

epidermal cells prior to visualisation microscopically. However akin to any type of

scientific technology there are limitations. Proteins with very short half-lives are

difficult to measure and proteins must be in enough quantity so as an adequate level

of detection is possible. Finally, experimental controls are performed. For example the

intensity and foci of fluorescence are monitored and checked to confirm that

fluorescence was due to localisation of a protein and not protein aggregation on the

microscope.

2.9.2 In vitro import of precursor proteins into

mitochondria

The procedures for in vitro imports were based on the methods described in (Whelan

et al., 1996). Import reactions used 180 μL of import master mix that was made up of:

2x import buffer, 1M MgCl2, 100 mM methionine, 100 mM ADP, 100mM ATP, 0.5 M

succinate, 0.5 M of fresh DTT, 0.5 M NADH and 100 mM GTP. Arabidopsis

mitochondrial imports were always carried out using freshly isolated mitochondria

from swirling liquid plant cultures. For a 4 lane import 100 μg of fresh mitochondria

were used with precursor protein (Whelan et al., 1996).

Protein imports were commenced by incubating the two reactions, one reaction with

the addition of Valinomycin (Val) and the other without at 25 °C for 20 min shaking at

350 rpm. The reactions were stopped by placing them on ice for 3 min. The reactions

containing no Val and Val were again divided into two. Proteinase K (PK) was added

Chapter 2. Materials and Methods

74

to one of the reactions with Val and one reaction without Val. All reactions were

incubated for a further 30 min on ice and 1μL of 100mM PMSF was added to the

reactions containing PK in order to inactivate the PK (Whelan et al., 1996). All

mitochondrial reactions were centrifuged for 3 min at 4°C at max speed to pellet the

mitochondria. The pellets were resuspended in 30 μL of 2x sample buffer, boiled for 3

min and run on a 16 % (v/v) SDS-PAGE gel (12 % (v/v) if the proteins were larger

than 20 kDa). The gels were coomassie stained for 2 hr, destained for 2hr, dried at 80

°C for 2h and exposed to a BAS TR2040 plate for 2 nights. The exposed plates were

read with the Personal Molecular Imager (Bio-Rad, Sydney, Australia).

2.9.3 In vitro import of precursor proteins into pea

chloroplasts

An import master mix was made prior to import commencement this contained: 1 x

import buffer, 100 mM MgCl2, 100 mM ATP, 100 mM Methionine, 1 M potassium

acetate and 1 M NaHCO3. Imports were always carried out using freshly isolated

chloroplasts, 50 μL of 500 μg chlorophyll/mL chloroplasts were added to each reaction

with 10 μL of precursor protein. The reactions were incubated for 25 min at 25 °C with

gentle agitation. The reaction was divided into two separate reactions, to one reaction,

1.5 μL of 5 mM CaCl2 and 2 mg/mL thermolysin was added. This reaction was

incubated on ice for 30 min and following incubation 2 μL of 0.5 M EDTA was added

to inactivate the thermolysin. Both reactions were centrifuged for 2 min at 830 g at 4

°C, the pellets were resuspended in 2 x sample buffer, boiled for 3 min and run on a

SDS-PAGE gel (Section 2.9.7 SDS-PAGE).

2.9.4 Mitoplast preparation following mitochondrial

in vitro imports

Following in vitro imports, outer mitochondrial membranes were ruptured via osmotic

swelling (Whelan et al., 1996). This produced mitoplasts that allowed the inner

Chapter 2. Materials and Methods

75

membrane and inter membrane space to become accessible to PK treatment.

Following in vitro imports mitochondria were pelleted by centrifugation at 20 000 g for

10 min at 4 °C. The supernatant was removed and the pellets were resuspended in

10 μL SEH buffer (250 mM sucrose, 1 mM EDTA, 10 mM Hepes-KOH, pH 7.4). To

this reaction 155 μL of 20 mM Hepes-KOH, pH 7.4 was added and left on ice for 15

min. The iso-osmotic conditions were restored by adding 25 μL of 2 M sucrose, to

release the inter membrane space proteins 10 μL of 3 M KCL was added. The

mitoplast samples were then divided into two, to which 50 μg of PK was added to one

reaction; both reactions were left on ice for 30 min. Following incubation 2 mM PMSF

was added to inhibit the PK activity. All mitoplast reactions were pelleted by

centrifugation at 20 000 g for 10 min at 4 °C, the pellets were resuspended in 2 x

sample buffer, boiled for 3 min and run on a 16 % (w/v) gel (12 % (w/v) if the proteins

were larger than 20 kDa) (Whelan et al., 1996). As with the in vitro import the gels

were coomassie stained, dried and exposed to a BAS TR2040 plate for 48 hand these

plates were read with the Personal Molecular Imager (Bio-Rad, Sydney, Australia).

2.9.5 Alkaline extractions

Alkaline extractions were performed following mitochondrial in vitro imports in order to

determine if proteins were membrane associated or if they were soluble (Terziyska et

al., 2005). Imports were carried out as described in Section 2.9.1 and the pellets

produced were resuspended in 0.1 M Na2CO3. Following incubation on ice for 10 min

the samples were centrifuged for 30 min at 20 000 g at 4 °C to separate the soluble

fraction in the supernatant from the insoluble fraction in the pellet. The soluble fraction

was treated with 10 % (w/v) TCA, heated to 65 °C to inactivate the protease,

incubated on ice for 10 min and centrifuged for 10 min at 20 000 g at 4 °C. The

soluble proteins contained in the pellet following this spin were resuspended in 1 x

Tris-glycine based running buffer and centrifuged for 10 min at 20 000 g at 4 °C. The

Chapter 2. Materials and Methods

76

insoluble fraction and the soluble fraction were both resuspended in 2 x sample buffer

and loaded onto an SDS-PAGE gel (Section 2.9.5).

2.9.6 Proteinase K titrations

Imports were carried out as described in Section 2.9.1 with increasing amounts of

proteinase K added. Six lanes of mitochondria and six lanes of mitoplasts had 0 μg,

2.4 μg, 4.8 μg, 9.6 μg, 19.2 μg and 38.4 μg of PK added. These samples were then

incubated on ice for 30 and following incubation, 1 μL of 100mM PMSF was added to

the reactions containing PK in order to inactivate the PK. All samples were centrifuged

for 3 min at 4 °C at max speed to pellet the mitochondria and mitoplasts. The pellets

were resuspended in 30 μL of 2x sample buffer, boiled for 3 min and run on a 16 %

(w/v) gel (12 % (w/v) if the proteins were larger than 20 kDa). The gels were

coomassie stained, dried and exposed to a BAS TR2040 plate for 48 h. The exposed

plates were read with the Personal Molecular Imager (Bio-Rad, Sydney, Australia).

2.9.7 SDS-PAGE

Polyacrylamide gels were used to run protein samples following import, alkaline

extractions and PK titrations (Lister et al., 2001). These were assembled using the

Bio-Rad PROTEAN IITM system (Bio-Rad, Sydney, Australia).Gels were typically

made of a 16 % (w/v) acrylamide separating component and a 4 % (w/v) acrylamide

stacking component. The separating gel was made with 375 mM Tris-HCL pH 8.8, 16

% (w/v) acrylamide and 0.1 % (w/v) SDS. This was degassed for 5 min prior to the

addition of 0.05 % (w/v) ammonium persulphate (AMPS) and 0.05 % (v/v) TEMED. To

resolve larger proteins on SDS gels 12 % (w/v) acrylamide was used in the separating

component. After the separating gel was set the stacking gel was prepared by adding

125 mM Tris-HCl pH 6.8, 4 % (w/v) acrylamide and 0.1 % (w/v) SDS. Similarly this

Chapter 2. Materials and Methods

77

solution was degassed for 5 min prior to the addition of 0.05 % (w/v) AMPS and 0.05

% (v/v) TEMED.

Gels were run in a discontinuous Tris-glycine buffer system consisting of; 25 mM Tris-

HCL pH 8.3, 200 mM glycine and1 % (w/v) SDS. The gel was run for 10 h at 10 mA

per gel limited to 200 V. Most gels were stained with coomassie stain, that consisted

of 0.25 %(w/v) Coomassie Brilliant blue R250, 40 % (w/v) ethanol and 10 % (v/v)

glacial acetic acid for 3 h. Gels that were not stained were used for Western blotting.

Following the staining, gels were destained in 30 % (v/v) ethanol, 12 % (v/v) glacial

acetic acid and 2% (v/v) glycerol for 2 h. Gels that contained radiolabelled proteins

were dried under a vacuum drier for 2 h at 80 °C using the Gel dryer 583 (Bio-Rad,

Sydney, Australia). The dried gels were carefully wrapped in cling wrap, exposed to a

BAS TR2040 plate for 2 nights in a film cassette and then the plate was read using

the Personal Molecular Imager (Bio-Rad, Sydney, Australia).

2.10 Western Blotting

2.10.1 Blotting with nitrocellulose membranes

Following the separation of proteins on Sodium dodecyl sulfate polyacrylamide gel

electrophoresis (SDS-PAGE), the gels were transferred to 0.45 μm supported hybond

C+ nitrocellulose membrane (Amersham, Sydney, Australia) (Jaeger and Wigge,

2007). The gels were equilibrated in ponceau (Appendix 1) for 1 h on a rocker whilst

10 sheets of Whatman® filter paper and 1 membrane were cut to the exact

measurements of the gel. The filter paper and membrane were both equilibrated in

transfer buffer for 10 min prior to transfer. Five sheets of Whatman® filter paper were

placed on the Phamacia (Amersham Pharmacia Biotech, Sydney, Australia) semi-dry

blotting apparatus, followed by the membrane, the gel, the remaining 5 sheets of filter

Chapter 2. Materials and Methods

78

paper and the cathode lid of the blotting apparatus. Gels were transferred for 1 h at

0.8 mA per cm2. Following transfer the membrane was quickly stained with

Ponceau-S stain (Appendix 1) for less than 1 min to confirm the locations of the

molecular weight markers and consistent transfer throughout the gel. The membrane

was destained in TBS-Tween (Appendix 1) and incubated in 1 % (v/v) blocking

solution from the Chemiluminescence Blotting substrate kit (BM, Sydney, Australia)

overnight at 4°C (Jaeger and Wigge, 2007).

2.10.2 Blotting with PVDF membranes

For proteins smaller than 20 kDa in size Immobilon-PSQ membranes were used. Gels

and Whatman® filter paper were equilibrated in Polyvinylidene fluoride (PVDF) transfer

buffer (Appendix 1) for 10 min whilst the membranes were washed in methanol for 15

min until they became opaque, the membranes were then rinsed in milli-Q water and

equilibrated in PVDF transfer buffer for 10 min. The filter papers, membranes and gels

were assembled and transferred on the Phamacia semi-dry blotter as described in

Section 2.10.1 and blocked overnight in 1 % (v/v) blocking solution prior to immune-

detection.

2.10.3 Immuno-detection

The UPOX1 antibody was expressed and purified by Dr Murcha in the Whelan lab and

provided for use in this thesis. This was produced using aa 1-137 of UPOX1, this

protein was then expressed in pdest15 fused to a GST tag. The GST tag was

removed using a Precision Protease leaving a 14 kDa protein for inoculation. The

protein was confirmed by mass spectrometry prior to injection into rabbit at 4 x 0.25

mg. The immuno detection experiments were blocked overnight, following this the

membranes were washed with TBS-Tween and incubated with primary antibody

diluted in 20 mL of TBS-Tween for 1 h with rocking. Following incubation with primary

Chapter 2. Materials and Methods

79

antibody the membranes were rinsed twice for 5 min and once for 15 min with TBS-

Tween. The membranes were incubated for 1 h in a 1 in 10 000 dilution of secondary

antibody (either anti-mouse or anti-rabbit) with rocking and then rinsed twice for 5 min

and once for 15 min with TBS-Tween.

Detection solution was made up using the substrates in the Chemiluminescence kit 30

min prior to use and equilibrated at room temperature. The 3 mL detection mixture of

1 % (v/v) substrate A in substrate B was poured onto the membrane, a plastic sheet

was placed underneath and above the membrane and all air bubbles were removed

prior to detection (Lister et al., 2007). Visualisation of the membrane was carried our

using the Image Quant RT ECL (GE Amersham Bioscience, Sydney, Australia)

according to manufacturer’s instructions.

2.11 BN PAGE

2.11.1 Preparation of BN PAGE gels

Prior to the preparation of Blue Native PAGE (BN-PAGE) gels, all solutions, casting

devices and pumps were chilled for at least 2 h at 4 °C. To prevent leakage of the 1.5

mm thick protein gel two pieces of Whatman® filter paper were placed under and 1

sheet of parafilm was placed above the rubber seal at the bottom of the glass plates.

Once the tubing is placed around the pump the injection needle is injected through the

bottom of the glass plates through the rubber seal. A pre-chilled light solution

(Appendix 1) was pumped through into the gel and once the level of the light solution

and heavy solutions (Appendix 1) were equal the valve for the heavy solution was

opened creating an increasing gradient higher in the gel. After the gel (in liquid form)

was fully injected between the two glass plates it was moved to room temperature to

speed the polymerization process which took approximately 1.5 h. After

Chapter 2. Materials and Methods

80

polymerization of the separating gel a 4 % (v/v) casting gel was poured on top with a

10 lane comb placed in it.

2.11.2 BN PAGE sample preparation

Digitonin was used to solubilise the mitochondrial samples for analysis by blue native

PAGE. Mitochondrial samples were resuspended in 5 % (w/v) digitonin extraction

buffer and incubated on ice for 20 min, 250 μg of protein was dissolved in 25 μL of

digitonin buffer with 1.25 mg digitonin. The samples were then centrifuged at 15 000 g

for 20 min at 4 °C and the supernatant containing the mitochondrial proteins were

again resuspended in 5 % (w/v) Serva Blue G prior to loading onto a blue native

PAGE gel.

To support the transfer of the protein into the gel a tension of 100 V was selected for

45 min with a variable current between 6 to 8 mA. For the separation of the proteins a

current of 15 mA was selected for 6 h with a limit to 500 V. After the gels completed

running they were washed with milliQ water and prepared for wet transfer.

2.11.3 Wet Transfer of BN PAGE gels

Prior to wet transfer the gel was soaked in transfer buffer for 30 min and PVDF

membrane cut to the exact dimensions of the gel were soaked in transfer buffer for

5 min along with two sheets of filter paper cut to the size of the sponges. The electro

blotting apparatus was assembled in transfer buffer by placing 1 sheet of filter paper

on top of the cathode cassette, followed by the gel, the membrane and the second

filter paper. All bubbles were rolled out with every layer and the cassette was placed

in the tank and electro blotted for 11 h at 50 mA. After transfer blue die was visible on

the membrane this indicated a good transfer, this was washed with methanol for 3

min. After marking the location of the complexes the membrane was immuno-detected

according to Section 2.10.3.

Chapter 2. Materials and Methods

81

2.12 Construction of a phylogenetic tree

Two phylogenetic trees of UPOX were created by using ClustalW2 and PANTHER

(Protein ANalysis THrough Evolutionary Relationships). ClustalW2 uses the

Neighbour-Joining method (Saitou and Nei, 1987). This algorithm functions by

clustering sequences by minimizing the sum of branch lengths. An output format that

ClustalW2 produces is the input format required by the tree-displaying program

FigTree v1.4.3. The PANTHER database was used for the second phylogenetic tree.

This resource is used for the classification of protein sequences by evolutionary

history, and by function. The database consists of protein-coding genes from 104

organisms that are classified by evolutionary relationships, by structured

representations of protein function including the Gene Ontology and biological

pathways. PANTHER phylogenetic trees show the evolutionary events including

speciation, gene duplication and horizontal gene transfer. Using these phylogenetic

trees orthologs, paralogs and xenologs are predicted. A hidden Markov model (HMM)

is constructed for each family and subfamily (Mi et al., 2016).

Chapter 3 Expression and regulation of UPOX1

82

Chapter 3

Expression and regulation of UPOX1

Chapter 3 Expression and regulation of UPOX1

83

3.1 Introduction

UPOX1 is a 104 amino acids long and is known as a highly stress-responsive gene

that was identified as one of the hallmarks of oxidative stress (Gadjev et al., 2006)

within the model plant Arabidopsis thaliana. To understand this gene in more depth

orthologs will be first examined. Two nuclear encoded genes have been identified as

exhibiting high levels of amino acids sequence identity to At2g21640 (UPOX1). These

genes have been named UPOX2 (At4g39235) and UPOX3 (At3g05570) (Uggalla,

2006). Gene duplication is a major evolutionary mechanism leading to the emergence

of new functions (Gu et al., 2004) and these genes may be a result of gene

duplication. In plant genomes the existence of multi-gene families are common and

one reason for maintaining these genes is that they have become functionally

specialized (Lynch and Conery, 2000). Genes within a family are known to exhibit

diverse transcriptional responses to oxidative stress (Dong and Adams, 2011). A

study by (Kim et al., 2005) identified that from 4222 duplicated gene pairs 22%

contained at least one member that was responsive to oxidative stress but from this

22% only 3% contained gene pairs that were both responsive to oxidative stress.

Possible orthologs of UPOX1 have been identified using BLAST (Uggalla, 2006).

Orthologs by definition (orthos meaning ’right’) are the same genes in a different

species that have evolved from a common ancestral gene by speciation, therefore

they should retain the same function. Paralogs on the other hand do not have any

functional connotation and the sequence similarity is the result of a gene duplication

that occurred after the speciation, in-paralogs are within a species whereas

out-paralogs are not (Fitch, 1970).

In this study we have analysed and compared how UPOX1 has diverged from UPOX2

and UPOX3 by examining their transcript expression patterns by performing qRT-PCR

and by using Genevestigator. In addition to these homologs, several other

Chapter 3 Expression and regulation of UPOX1

84

mitochondrial genes that are induced by treatments that perturb mitochondrial function

were also examined in this study.

Microarray analysis suggests that UPOX1 may be co-expressed with AOX1a and

NDB2 (Clifton et al., 2005), however, there is no data that shows the transcript profiles

of these genes in response to stress treatments over a time period. Therefore qRT-

PCR analysis was also conducted in order to observe the alterations in transcript

abundance during a time course for AOX1a, NDB2, AtOM66, UBC (AT5G25760) and

PR1 in Col-0 plants and in suspension cell cultures.

To further examine possible pathways that regulate mitochondrial stress response at

a transcriptional level the transcript abundances of UPOX1 and various other nuclear

encoded mitochondrial proteins known to be stress responsive were analysed in

defense signalling mutants and Col-0 plants.

Complex regulatory networks are formed from the interactions of different transcription

factors influencing the expression of genes in response to various internal and

external stimuli (Elkon et al., 2003). In addition to the cis-acting elements present

upstream of the transcriptional start site, it has been documented that 5’ introns can

also influence the level of expression at many levels including initiation of

transcription, mRNA metabolism, mRNA editing and translation. In order to obtain a

comprehensive understanding of the function of UPOX1 an investigation into the

induction of UPOX1 at a molecular level is required. Therefore, the upstream region of

UPOX1 was studied in detail and a set of potential regulatory elements were

functionally characterised as UPOX1 has a 5' intron 477 bp upstream of the

translational start site.

3.2 Aims and Strategies

Chapter 3 Expression and regulation of UPOX1

85

The specific aim of the research described in this chapter is to develop a

comprehensive understanding of the transcript expression pattern and regulation of

UPOX1.

This will be conducted by studying;

- The transcript expression kinetics of UPOX1 under various stress conditions

- The transcript expression profile in various mutants

- Promoter- GUS histochemical staining of plants

- The promoter region of UPOX1 via biolistic transformation using bioinformatic

tools

3.3 Results

3.3.1 Comparisons between the UPOX1 family

As UPOX2 and UPOX3 display high levels of sequence identity to UPOX1, the protein

and DNA coding sequences for these genes were investigated. Nucleic acid and

protein sequences were obtained from TAIR (Swarbreck et al., 2007) and multiple

sequence alignments were performed using ClustalW2 (Larkin et al., 2007). This

analysis revealed that only the N terminal region differentiates UPOX1 from UPOX2

and UPOX3 as the C terminal regions are highly conserved amongst all three genes.

Figure 3.1 A shows the entire 104 amino acid length of UPOX1. UPOX1 appears to

have additional amino acids at the N terminal region and it is thought that this is a

characteristic of mitochondrial targeted sequences (Glaser et al., 1998). Their

deduced amino acid sequences show 76% sequence identity between UPOX2 and

UPOX3 and 52% sequence identity when UPOX2 and UPOX3 are compared with

UPOX1 (Figure 3.1 B). The coding sequences of UPOX2 and UPOX3 have 80%

sequence identity between them and they both have 75% sequence identity with

UPOX1 (Figure 3.1 C).

Chapter 3 Expression and regulation of UPOX1

86

3.3.2 Construction of a phylogenetic tree

Orthologs of UPOX1 were identified using two independent applications. The first

phylogenetic tree (Figure 3.2 A) was generated by ClustalW2 and displayed using

FigTree v1.4.3 following the use of Basic Local Alignment Search Tool (BLAST). The

second phylogenetic tree (Figure 3.2 B) was generated using PANTER (Protein

ANalysis THrough Evolutionary Relationships) databases (Mi et al., 2016).. Analysis

of the phylogenetic tree created through the PANTHER database reveals more

orthologous relationships than that using ClustalW. Analysis of this second tree

reveals that there are seven orthologs of UPOX1 in Oryza sativa, one in Sorghum

bicolor, one in Brachypodium distachyon, five in Glycine max, three orthologs in

Populus trichocarpa, two in Solanum lycopersicum and one orthologs in

Physcomitrella patens. It is also apparent that UPOX2 and UPOX3 may be related

closer to each other than they are to UPOX1. Interestingly upon further investigations

using SUBA3 a subcellular localisation tool for Arabidopsis (Tanz et al., 2012; Hooper

et al., 2014) the predicted in silico subcellular localisation of UPOX2 and UPOX3 were

found to be in both the nucleus and cytosol whereas UPOX1 had mixed predictions

with 2 out of 12 predictors suggesting mitochondrial localisation. There were further in

silico localisations available for the orthologs in Oryza sativa and these were localised

to multiple subcellular localisation including the chloroplast, cytosol and golgi.

3.3.3 Establishment and optimisation of qRT-PCR assays

The full length coding sequences for UPOX1, UPOX2 and UPOX3 were amplified and

qRT-PCR primers (Appendix 2) were designed for each gene as per Section 2.7. The

oligonucleotides were 16 to 20 base pairs (bp) in length with GC contents ranging

from 47% to 62% and melting temperatures ranging from 50 °C to 70 °C. The

sequence identity between UPOX1, UPOX2 and UPOX3 was high and therefore the

primers were designed in the regions that displayed lower sequence identity in order

to obtain gene specific primers sets.

Chapter 3 Expression and regulation of UPOX1

87

Quantitative RT-PCR standards were generated for UPOX1, UPOX2 and UPOX3.

The full length expected coding sequence was used and the sizes for UPOX1,

UPOX2 and UPOX3 were 315 bp, 261 bp and 273 bp respectively. The standards

were quantitated using PicoGreen® and diluted to produce a master stock at 1 mol/

L. Serial dilutions of this master stock were used as a standard in all qRT-PCR

assays. The qRT-PCR reactions were optimised for linearity and specificity. Linearity

is essential for all qRT-PCR assays as this determines the accuracy with which

samples of unknown concentration are determined. A four point standard curve was

utilised for all the assays conducted and qRT-PCR efficiencies of greater than 95%

were maintained for all assays performed (Giraud et al., 2008).

Cross reactivity assays were also performed using the Roche Light Cycler® software

and melt peak charts according to Section 2.7.5 which confirmed that the primers

were specific for the target transcript.

3.3.4 Quantitative RT-PCR assays for UPOX1, UPOX2 and

UPOX3 in suspension cell culture

Transcript levels of UPOX genes were monitored in Arabidopsis cell culture lines

treated with a variety of chemicals. A range of chemical treatments were selected for

their broad impact on cellular function or for their ability to inhibit a specific

mitochondrial or cellular process. The cell cultures used for the qRT-PCR assays

were isolated from three biological replicates, from each of these, two technical

replicates were isolated. From 120 ml flasks grown under the conditions in Section

2.2.1 a 5 mL aliquot was removed at the 0 h time point prior to the addition of any

chemicals. A range of chemical treatments were selected for their broad impact on

cellular function or for their ability to inhibit a specific mitochondrial or cellular process.

Following the addition of stress treatments 5 ml samples were taken, at 3 h, 12 h

and 24 h after the stress treatment was applied. These samples were then vacuum

filtered and snap frozen. From this, the RNA was isolated and cDNA was synthesized

Chapter 3 Expression and regulation of UPOX1

88

according to (Clifton et al., 2005). Following this, qRT-PCR assays (using the primers

in Appendix 2) were conducted and the results were normalized to ubiquitin (UBC) as

a housekeeping gene (Clifton et al., 2005; Czechowski et al., 2005). The stress

treatments used included citrate, hydrogen peroxide (H2O2), rotenone and salicylic

acid. Citrate is a TCA metabolite and is known to disturb metabolic homeostasis

(Vanlerberghe et al., 2002). H2O2 is a ROS signal and an agent for cellular damage

(Maxwell et al., 2002). Rotenone inhibits complex I and thus affects the functioning on

the mitochondria (Li et al., 2003). Lastly, salicylic acid is an un-coupler of respiration

and is a common defense signalling hormone .

The qRT-PCR assays in Figure 3.3 demonstrate that the transcript abundance of

UPOX1, UPOX2 and UPOX3 increase with these treatments. However, there were

differences in the magnitude and kinetics of their response. In the case of UPOX1 the

response to all treatments except salicylic acid peaked 24 h after stress treatment;

salicylic acid produced a peak at the 12 h time point with transcript abundance

increasing approximately 25-fold. At the 24 h time point there were significant

increases in the transcript abundance in response to H2O2, rotenone and salicylic

acid. The changes in transcript abundance corresponded to-5 fold, 20-fold and 5-fold

increase respectively. The increase in transcript abundance for UPOX2 and UPOX3

were of a significantly lesser magnitude than that of UPOX1. However there was still a

significant increase in transcript abundance in response to salicylic acid for UPOX2.

The response peaked at the 3 h time point with transcript abundance increasing 7-

fold, this was maintained at the 12 h time point and at the 24 h time point a slight

reduction in transcript abundance was measured that corresponded to a 6-fold

increase. The transcript abundance of UPOX3 in response to citrate, H2O2, rotenone

and salicylic acid did not increase above 3.5-fold and the only significant changes

observed were in response to citrate. This change in transcript abundance showed a

Chapter 3 Expression and regulation of UPOX1

89

3-fold increase at the 12 h time point and peaked with a 3.5 fold increase at the 24 h

time point.

3.3.5 Transcript kinetics of nuclear genes encoding

mitochondrial proteins

The results shown in Figure 3.4 were produced using the same suspension cell

cultures used to produce the results for Figure 3.3. This permits the expression

kinetics of UPOX1, UPOX2 and UPOX3 to be compared to the results obtained in

Figure 3.4. Following micro array analysis it was observed that the expression of

AOX1a was co-expressed with NDB2 and UPOX1 (Figure 3.6). To examine the

alterations in transcript abundance of several nuclear genes encoding mitochondrial

proteins in response to stress treatments that inhibit specific mitochondrial or cellular

processes qRT-PCR analysis was conducted; the genes tested were AOX1a, NDB2,

AtOM66, UBC and PR1. UBC (AT5G25760) was used as a housekeeping gene as it’s

known to be unresponsive to a wide range of treatments and PR1 is a pathogen

related protein 1a and is a positive control for a response to SA (Clifton et al., 2005;

Czechowski et al., 2005).

The qRT-PCR analysis demonstrated the transcript abundance for AOX1a in

response to H2O2, rotenone and SA all peaked at the 3 h time point. The transcript

abundance increased greater than 6-fold and at the consequent 12 h and 24 h time

points the transcript abundance of AOX1a in response to all the treatments decreased

whilst maintaining a higher abundance than the levels observed at the 0 h time point.

The transcript profile of NDB2 most resembles that of AOX1a, however, there were

distinct differences. The transcript abundance of NDB2 peaked at the 12 h time point,

an approximate 4-fold increase was observed in response to H2O2 and SA whilst an 8-

fold induction was observed in response to rotenone treatment. NDB2 was the only

gene to significantly increase in transcript abundance in response to citrate with a

Chapter 3 Expression and regulation of UPOX1

90

relatively modest 2-fold increase observed at the 24 h time point. The transcript

profiles of AOX1a and NDB2 were similar in the aspect that the transcript abundance

of both genes decreased at the 24 h time point. The expression profile of AtOM66

differed greatly as its response to SA produced the greatest increase in transcript

abundance at all time points after the 0 h time point. At the 12 h time point a 30-fold

increase was observed and at the 24 h time point a 25-fold increase was observed in

response to SA. The transcript abundance of AtOM66 in response to citrate and H2O2

treatments produced inductions below 5-fold at all 4 time points whilst the response to

rotenone was slightly higher than 5 fold for the 12h and 24 h time points. The

transcript abundance of PR1 was as expected with only SA producing a significant

change in transcript abundance with a greater than 140-fold induction observed at the

24 h time point.

3.3.6 Transcript analysis in response to UV stress

To further investigate the response of UPOX1 and AOX1a in Col-0 plants to

environmental stress qRT-PCR analysis was conducted in response to UV stress

(Figure 3.5). Arabidopsis water cultures were monitored 6 h following 5 min treatment

of UV-C according to Section 2.2.5. For each sample, total RNA was isolated from

three pools of tissue, each consisting of three independent pots. The qRT-PCR

analysis demonstrated the transcript abundance for UPOX1 and AOX1a increased

significantly following UV stress.

3.3.7 In silico response of UPOX1, UPOX2 and UPOX3 to

a wide range of stimuli and mutations

Within the Genevestigator database the Response Viewer tool was used to extract the

relative expression data as fold changes for UPOX1, UPOX2, UPOX3, AOX1a, NDB2,

AtOM66, PR1 and UBC (Hruz et al., 2008). This was viewed as a heat map in

response to stimuli and mutations, the 50 most up-regulated and down-regulated

Chapter 3 Expression and regulation of UPOX1

91

stimuli and mutations can be seen in Figure 3.6. Interestingly among the top 50 stimuli

that up regulate the expression of UPOX1, 17 were chemical treatments, 14 were

stress treatments, 10 were light treatments, 4 were biotic treatments, 3 were nutrients,

1 was a hormone and the 1 remaining was a cell sorting and protoplasting stimulus.

The stimuli that produced UPOX1 to be most down regulated were represented by 12

stress treatments, 12 nutrients, 9 chemicals, 7 hormones, 3 light treatments, 2

temperature treatments, 2 biotic treatments, 1 elicitor, 1 photoperiodic shift and 1

programmed cell death stimulus. It is also clear that the expression of UPOX2 and

UPOX3 to these stimuli did not result in a change in transcript abundance to the

degree shown by UPOX1. From the top 50 stimuli that up regulate UPOX1, 46 (92 %)

of these stimuli also up regulated AOX1a. In addition to this, 70% of these 50 stimuli

also caused an up regulation of NDB2 and AtOM66. Furthermore from the top 50

stimuli that caused a down regulation in UPOX1 greater than 50 % of these stimuli

also produced a down regulation of AOX1a.

3.3.8 Mitochondrial genes induced under stress in

signalling mutant backgrounds

By observing the expression patterns of UPOX1, UPOX2, UPOX3, AOX1a, NDB2,

AtOM66, PR1 and UBC in various mutants it can assist in characterizing the pathways

that regulate their expression (Figure 3.6). There were significant changes in the

expression of UPOX1 in several mutant lines. UPOX1 was most up-regulated in the

flu.2 mutants and this mutant is a negative regulator of impaired chlorophyll

biosynthesis and results in ROS production when shifted from dark to light

(Meskauskiene et al., 2001). The mutant in which UPOX1 was second most up-

regulated was the ang4 (Berná et al., 1999; Robles and Micol, 2001) mutant, this

mutant increased cell cycle duration in young leaves. The third mutant that UPOX1

was most up-regulated in was the csn5 mutant (Dohmann et al., 2005), these mutants

Chapter 3 Expression and regulation of UPOX1

92

have a characteristic phenotype of photo-morphogenic growth in the dark and

post-germination growth arrest. The flu and csn mutants comprise the top 7 mutants

that most upregulate UPOX1. This provides a clue as to the pathways that UPOX1

could be functioning in as it may be related to these mutants. Furthermore of the top

50 mutants that caused UPOX1 to be up regulated, the top 16 mutants that most up

regulated UPOX1 also produced an up regulation of AOX1a, NDB2 and AtOM66. An

additional 15 of the 50 mutants also produced an up regulation in the transcript levels

of AOX1a, NDB2 and AtOM66.

To investigate the signalling pathways that regulate UPOX1, UPOX1 expression

levels were assessed in several additional mutant lines compared to Col-0 plants as

shown in Figure 3.7. The transcript abundance of several genes encoding

mitochondrial proteins was also assessed in a variety of mutant lines. The genes

tested alongside UPOX1 included a highly stress responsive alternative oxidase

AOX1a (Yu et al., 2001), AtOM66 that encodes a protein that is present in a homo-

multimeric protein complex on the outer mitochondrial membrane and plays a role in

cell death and amplifying salicylic acid signalling (Zhang et al., 2014), a gene that

encodes an external NAD(P)H dehydrogenase NDB2 that follows a similar pattern of

transcript expression to AOX1a (Clifton et al., 2005), a pathogen related protein that

was used as a positive control for salicylic acid (SA) PR1a (Shah, 2003) and a house

keeping gene that is unresponsive to a wide range of treatments UBC (Czechowski et

al., 2005). The qRT-PCR for AOX1a and NDB2 were conducted in a collaboration

within the Whelan lab by Dr Giraud and Dr Ho. The mutant lines chosen were all

affected in phytohormone signal transduction pathways. NahG mutant plants break

down SA by expressing bacterial salicylate hydroxylase (Lawton et al., 1995; Bowling

et al., 1997), pad4 is a phytoalexin deficient mutant that is essential for the SA

dependent defense pathways (Glazebrook and Ausubel, 1994; Glazebrook et al.,

2003), eds1 is an enhanced disease susceptible mutant (Falk et al., 1999) and npr1

Chapter 3 Expression and regulation of UPOX1

93

mutants are non-expressers of the gene PR1 which is required for the expression of

defense genes (Cao et al., 1994; Shah, 2003). Pad 4 mutants act upstream of SA

whereas eds1 mutants and npr1 mutants act downstream of SA. The other mutant

lines chosen were the jar1 mutant line that are associated with insect and

necrotrophic defense and is deficient in the jasmonic acid signalling pathway

(Staswick et al., 1992), the abi3 line that interacts with the abscisic acid (ABA)

responsive transcription factors by encoding a viviparous like transcription factor

(Giraudat et al., 1992) and the etr1 mutant line that confers ethylene insensitivity in

Arabidopsis (Chang et al., 1993).

In NahG plants relative to Col-0 plants it was observed that the transcript abundance

of AtOM66 and PR1 had reduced by approximately 100-fold, whereas there was only

a 2.5 fold reduction of AOX1a and 5-fold reduction in transcript abundance of NDB2

and UPOX1 (Figure 3.7). In the pad4 and eds1 mutant lines the transcript abundance

of AOX1a differed greatly to that of NDB2 and UPOX1, there was a greater than 2-fold

increase in AOX1a transcript abundance whilst the transcript abundance of NDB2,

UPOX1 and AtOM66 resulted an approximate 5-fold reduction in the pad4 plants. In

the eds1 line there was a slight increase in the transcript abundance of UPOX1, a 5-

fold decrease in AtOM66 and the transcript abundance of NDB2 remained

unchanged. The expression of PR1 in the pad4, eds1, npr1 and jar1 plants was

similar to that expressed in the NahG plants, the transcript abundance was only just

detectable and it was approximately 100-fold reduced relative to Col-0 plants, the only

significant increase in abundance of PR1 was observed in the abi3 plants. The

expression pattern of AtOM66 clearly differs from that of AOX1a, NDB2 and UPOX1

in the npr1, jar1, abi3 and etr1 plants. From its induction pattern AtOM66 is consistent

with a role for SA signal induction as its transcript abundance decreases in NahG,

pad4, eds1 plants. However it distinguishes itself from PR1 by showing that it is not

dependent on NPR1. The expression patterns of AOX1a, NDB2 and UPOX1 were

Chapter 3 Expression and regulation of UPOX1

94

similar in the npr1, jar1, abi3 and etr1 lines, the transcript abundance of all these

genes increased greater than 2-fold (Ho et al., 2008).

3.3.9 GUS histochemical staining

GUS histochemical analysis is another method to confirm and localise the expression

of genes of interest. The upstream 1.5 kb region of the UPOX1 promoter was fused to

β-glucuronidase and transformed into Arabidopsis according to the methods detailed

in Skirycz et al., (2010). This line was obtained in collaboration with Dr Van Aken and

Inge De Clercq in the lab of Prof. Van Breusegem (VIB Ugent, Belgium). The GUS

activities in the whole root tip, vascular tissue, the siliques and mature flowers were

assayed by myself. Figure 3.8 shows 6 images and the blue regions indicate the

degree to which the GUS has stained the plant material. The root tip, the base ends of

the silique and the mature flowers reveal strong GUS staining and the localisation of

this signifies the localisation of UPOX1 expression in Arabidopsis plants. Overall, the

UPOX1 promoter appears to be active in a variety of tissues throughout the plant.

3.3.10 Identification of putative sequence elements in

the upstream region of UPOX1

To obtain a better insight in how the stress-responsive expression of UPOX1 is

regulated, the UPOX1 promoter was analysed and searched for the presence of

regulatory elements. In total 14 putative regulatory cis-sequence elements were

identified in the upstream region of UPOX1 using five prediction programs; Plant Care

(Rombauts et al., 1999), PLACE signal scan (Higo et al., 1999), AthaMap (Steffens et

al., 2004), Athena (O'Connor et al., 2005), and AGRIS (Palaniswamy et al., 2006).

Of these 14 elements, there was 1 MYB motif, 1 I and B overlapping elements which

were found to be functional in AOX1a (Clifton et al., 2005), there were 2 WRKY

elements both in the forward orientation, these elements have been found to be

Chapter 3 Expression and regulation of UPOX1

95

involved in stress and defense signalling (Eulgem and Somssich, 2007). Also present

were 2 RAV1 elements, the RAV1 elements in pepper have been identified as having

a role in biotic and abiotic stress resistance (Sohn et al., 2006) and there were 6

elements that are known as stress responsive elements, these elements are highly

over represented in stress responsive genes (Figure 3.9). Furthermore a

CTTGGCCACG motif was identified that is known to be overrepresented in genes

responding to mitochondrial dysfunction and is bound by NAC-type transcription

factors (Van Aken and Whelan, 2012; De Clercq et al., 2013; Ng et al., 2014).

.

3.3.11 Functional analysis of the B and I elements in the

upstream region of UPOX1

The B and I element had been previously identified to be functional in AOX1a and

NDB2 genes (Ho et al., 2008). These genes are co-expressed with UPOX1 and

therefore the functionality of these elements were investigated in UPOX1 (Figure

3.10). In order to test sequence elements for functionality, the 1100 bp upstream

region of UPOX1 was cloned into the pLUS vector (Ho et al., 2008). The pLUS vector

was created so promoters could be analysed via biolistic transformation. The

promoter of interest was fused to the glucuronidase reporter gene, and a

35S:luciferase gene was used for normalisation as transformation control.

Biolistic transformations were performed in suspension cell cultures that were

maintained as described in Section 2.2.1 and 4 week old Arabidopsis Col-0 leaves

maintained as described in Section 2.2.3. The I and B sequence elements were

mutated from a wild type promoter via site directed mutagenesis as described in

Section 2.4.7. Following the individual deletion of the I and B elements a construct

was also created that had both the I and B elements mutated. The effect of the

deletion of the sequence elements on the promoter’s ability to drive GUS expression

under stress treatments was also tested in both suspension cell cultures and Col-0

Chapter 3 Expression and regulation of UPOX1

96

leaves. The stress treatments used were H2O2 and rotenone. The GUS activity of the

wild type promoter under no stress treatment was set to 100% and the activity for the

deletions under mock (water/ethanol) treatments and stress treatments were

expressed relative to this.

(Rosner, 2006). The t-test assuming unequal variance was used to determine the

statistical difference in endogenous levels of GUS activity observed between the wild

type promoter and the mutated promoter within a treatment and also the statistical

difference in GUS activity between mock treated samples versus treated sample

within a promoter construct, in all situations the significance level was set at p ≤ 0.05.

In untreated suspension cell cultures the deletion of the I element from the upstream

region of UPOX1 resulted in a loss of GUS activity by ~40%. The deletion of the B

and the I+B combination elements resulted in a loss of GUS activity by ~50%. For the

wild type region, the mutated I element region and the mutated B element region the

addition of H2O2 produced an approximate doubling in GUS activity. Following the

deletion of the I+B element the increase observed following the addition of H2O2 was

eliminated. No significant responses to rotenone treatment were observed for the wild

type region or the mutated regions.

In untreated 4 week old Arabidopsis leaves the deletion of the I element and the I+B

element from the upstream region of UPOX1 resulted in a loss of GUS activity by

~75% under control conditions. The deletion of the B element resulted in a loss of

GUS activity by ~50%. For the wild type region, the addition of H2O2 produced an

increase in GUS activity by ~ 70%, however, following the deletion of the I, B and I+B

elements this induction following the addition of H2O2 was eliminated. There was no

significant response to rotenone treatment observed in the wild type region or the

mutated B element region, however, following the addition of rotenone to the mutated

I and I+B element regions there was an ~50% reduction in GUS activity.

Chapter 3 Expression and regulation of UPOX1

97

From the results obtained from the suspension cell assays it appears that all three

elements, I, B and I+B are positive regulators, as they show a decrease in GUS

activity when deleted. They appear to be positive regulators under untreated

conditions and following the treatment of H2O2. The result obtained from the 4 week

old Arabidopsis leaves concurs with the results obtained in the suspension cell assays

apart from one aspect. The assays in the 4 week old Arabidopsis leaves show that the

addition of rotenone in the mutated I and I+B element regions also appears to be a

negative regulator.

3.4 Discussion

A comprehensive study on the transcript expression kinetics of UPOX1 and its

homologs, UPOX2 and UPOX3, was conducted. This study incorporated qRT-PCR

analysis, promoter activity and microarray data analysis. This study established that

the expression profiles of these homologs differed considerably. This unique pattern

of transcript expression could suggest that the regulatory mechanisms controlling the

expression of each of these genes are largely independent and unrelated. This

conclusion correlates with previously conducted research which has established that

expression diversity is a means by which genes are evolutionarily retained (Yanhui et

al., 2006; Dong and Adams, 2011).

Overall the qRT-PCR assays in Figure 3.3 showed that UPOX1 is significantly more

stress responsive than its homologs UPOX2 or UPOX3. Furthermore, this analysis

revealed the kinetics of its stress response. The expression levels for UPOX1 peaked

at the 24 h time point. This indicates that the stress treatments used (citrate, H2O2,

rotenone and SA) likely cause a secondary stress response in UPOX1.

Chapter 3 Expression and regulation of UPOX1

98

Genevestigator analysis illustrated that the expression profile of UPOX1 in response

to various stress treatments and in various mutant lines differs from the expression

profiles of UPOX2 or UPOX3. This evidence correlates with the hypothesis that genes

within a family specialize and exhibit differing expression profiles.

Investigations into the transcript abundances of UPOX1 compared to that of AOX1a,

NDB2 and AtOM66 in response to external stimuli and in mutant backgrounds found a

high degree of correlation between UPOX1, AOX1a, NDB2 and AtOM66 and in

particular between UPOX1 and AOX1a. This suggests that there may be an overlap in

the pathways responsible for the induction of these genes.

Further qRT-PCR assays allowed the transcript abundance of UPOX1 to be

compared to AOX1a, NDB2, AtOM66 and PR1. The pattern of response of AOX1a

and NDB2 revealed that they did not co-ordinate with UPOX1. This was important as

the transcript abundance changes of AOX1a has been previously characterised as

important in the typical mitochondrial stress response pathway (Rhoads et al., 2006;

Suzuki et al., 2012). The difference in the transcript responses indicates involvement

in a potentially uncharacterised signal transduction pathway(s) regulating gene

expression for proteins that function in mitochondria.

An alternative approach was taken to further characterise the pathways that regulate

the expression of mitochondrial targeted stress responsive proteins by observing their

transcript abundance in defense signalling mutants. It was found that there were

distinctive pathways that regulate mitochondrial stress response at a transcriptional

level for UPOX1, AOX1a, NDB2 and AtOM66. This was first evident in the NahG

plants: UPOX1, AOX1a and NDB2 transcript abundance were reduced by

approximately 5-fold, however, AtOM66 expression was reduced greater than 100-

fold. This reveals that there were two distinct modes of action of SA on these

transcripts. AtOM66 was highly responsive to the application of SA and indicates a

Chapter 3 Expression and regulation of UPOX1

99

direct effect on transcription initiation. Conversely from the analysis of the promoters

and transcripts in this study it is proposed that SA acts to increase transcript

abundance for AOX1a, NDB2, and UPOX via a posttranscriptional mechanism. There

is further support for this hypothesis as a similar response was observed from

UPOX1, AOX1a and NDB2 in the etr1, abi3, jar1 and npr1 mutant lines, again

differing from the expression of AtOM66. Finally it was clear that the transcript

abundance of only AOX1a increased in the pad4 and eds1 mutant plants, this

indicates a separate regulatory pathway is involved for AOX1a and the induction of

AOX1a is SA independent in these lines. However from this study alone it is

impossible to conclude the exact effects of these mutants on altering transcript

abundances and it must be acknowledged that other factors could be acting in

conjunction with the effects of these mutant lines as the interactions between various

phytohormone signalling pathways are very complex (Erb et al., 2012).

To determine if UPOX1 was not only co-expressed with AOX1a but also co-regulated,

two elements previously found to be functional in AOX1a were assayed for

functionality within the upstream region of UPOX1. Elements I and B largely overlap

with a previously identified potential binding site for the transcription factor Abscisic

acid insensitive 4 and were tested independently and together. It was found that in all

three cases they acted as positive regulators. In addition the response of the UPOX1

promoter to H2O2 was consistent with it being defined as a marker of oxidative stress

(Gadjev et al., 2006). However as UPOX1 promoter activity was largely unresponsive

to rotenone and this differed considerably to the promoter activity of AOX1a (Ho et al.,

2008) it must be inferred that there are differences present in the nature of the signals.

In recent publications it has been found that the palindromic overlapped region of

element I+B has been called the NAC binding site (NACBS) and this has been found

to bind both ANAC017 and ANAC013 (Van Aken and Whelan, 2012; De Clercq et al.,

2013; Ng et al., 2014).

Chapter 3 Expression and regulation of UPOX1

100

In conclusion the use of transcript analysis, defense signalling mutants and promoter

activity allows for a thorough study of the intricate mechanisms underlying gene

expression under stress conditions. Through this research and following from what is

presented in (Ho et al., 2008) it can be concluded that there were 3 general pathways

that mediate mitochondrial stress response at the transcriptional level. The first is a

SA dependent pathway represented by AtOM66, therefore this implies that only

AtOM66 is regulated at a transcriptional level whereas UPOX1, AOX1a and NDB2 are

regulated at a post-transcriptional level. The second is illustrated by the lack of

response by UPOX1 to rotenone which differs significantly to AOX1a. This implies that

a set of different transcriptional factors are involved in the targeting of CARES by

H2O2 and rotenone. The third pathway is revealed by AOX1a as its expression differs

from UPOX1 and NDB2, this suggests a different pathway involved via EDS1 and

PAD4 to regulate only AOX1a. From the transcript analysis performed as part of this

study and further investigations of the promoter elements (Ho et al., 2008) it can also

be concluded that SA acts to increase transcript abundance for UPOX1, AOX1a and

NDB2 via a post-transcriptional mechanism, in contrast to AtOM66 which is under

direct transcriptional control. It can also be concluded that although H2O2 and

rotenone can both act transcriptionally and their CAREs overlap there is a distinction

in the response mechanism of UPOX1 compared to the other genes tested. The fact

that UPOX1 shows a peak in response to rotenone at the 24 h time point is the

evidence for the different transcription factors being involved, this is diagrammatically

represented in Figure 3.11.

Chapter 3 Expression and regulation of UPOX1

101

At4g39235 ---------METQTNQS---PKPAM--TSCRKKVKDD-ATFFEDVKDHIDDFIHASMDEH 45

At3g05570 ---------METQKTQAENPPKPAT--SSCRKKTKDD-ANFLEDVKDHIDDFINASMDDH 48

At2g21640 MASQTNQTVVDTKKIETENPPKPQVPASSCRKRVKDDNATFFANLKDHMDEFIHASMDEH 60

::*:. :: *** :****:.*** *.*: ::***:*:**:****:*

At4g39235 KSCFQKTIKKMFG---LSKAVAEKQAVEAKG-VESQLPLQTTVSE 86

At3g05570 KNCFNKTIKKMFG---LSKAVADKQQSEAKGGVESYLPLQTTVSD 90

At2g21640 KTCFKNTMDKIFGSFSKAEAVAEKQ-IEAKEVVEIHSPLQTAVTK 104

*.**::*:.*:** ::***:** *** ** ****:*:.

UPOX1 UPOX2 UPOX3

UPOX3 75 80 100

UPOX2 75 100

UPOX1 100

UPOX1 UPOX2 UPOX3

UPOX3 52 76 100

UPOX2 55 100

UPOX1 100

A)

B) C)

UPOX 1

UPOX 2

UPOX 3

UPOX 1

UPOX 2

UPOX 3

Figure 3.1 Comparisons of UPOX1, UPOX2 and UPOX3 sequences.

A) A ClustalW2 multiple sequence alignment of UPOX1, UPOX2 and UPOX3 protein

coding sequences. B) Percentage sequence identity between protein sequences.

C) Percentage sequence identity between nucleic acid sequences.

The colours represent the physicochemical propeties of the protein residues. (small

and small hydrophobic residues are depicted in red, acidic residues are blue, basic

residues are magenta, hydroxyl/ sulfhydryl/ amine are in green and other unusal

amino/ imino acids are in grey). An * (asterisk) indicates positions which have a

single, fully conserved residue. A : (colon) indicates conservation between groups of

strongly similar properties - scoring > 0.5 in the Gonnet PAM 250 matrix. A . (period)

indicates conservation between groups of weakly similar properties - scoring =< 0.5

in the Gonnet PAM 250 matrix.

Chapter 3 Expression and regulation of UPOX1

102

A)

Fig

ure

3.2

A)

UP

OX

1o

rth

olo

gu

es

.(A

)P

hylo

gene

tic

tre

ein

ferr

ed

from

asim

ultane

ous

com

pa

riso

nofU

PO

X1

ort

holo

gues

based

on

se

quen

ce

sim

ilari

ty.T

he

tree

was

con

str

ucte

dusin

gC

lusta

lW2

an

dF

igT

ree

v1.4

.3.

PO

PT

R_

000

5s22720

Pop

ulu

str

ich

ocarp

a

PO

PT

R_

000

4s16280

Po

pu

lus

tric

ho

carp

a

OS

03

G0

74555

0O

ryza

sativa

UPOX3At3g05570

Ara

bid

opsis

Thalia

na

UPOX2(A

t4g3

92

35

)A

rabid

op

sis

Tha

liana

PH

YP

AD

RA

FT

_1

9838

Physcom

itre

llapate

ns

UPOX1(At2g21640)

Ara

bid

op

sis

Th

alia

na

Os03g53390

Ory

za

Sativa

Os03

0724

600

Ory

za

Sa

tiva

0.4

5

0.3

30

.24

5

0.2

50.0

73

0.0

41

0.0

99

0.1

34

0.1

52

0.0

73

0.0

67

0.0

15

0.0

21

0.0

41

0.0

43

Chapter 3 Expression and regulation of UPOX1

103

Embryophyta

Magnoliophyta

Poaceae

Poaceae

Poaceae

BEP_clade

AN16

BEP_clade

Os03g53390 Oryza sativa

Os030724600 Oryza sativa

OS03G0745550 Oryza sativa

Os03g51470 Oryza sativa

Sb01g009650 Sorghum bicolor

BEP_clade

AN13

OJ1499_D04.6 Oryza sativa

BRADI1G10250 Brachypodium distachyon

OsJ_26398 Oryza sativa

Os08g0204632 Oryza sativa

Sb01g009660 Sorghum bicolor

Poaceae

Pentapetalae

rosidsfabids

AN47GLYMA11G07305 Glycine max

GLYMA04G16401 Glycine max

GLYMA01G38260 Glycine max

GLYMA06G46580 Glycine max

GLYMA02G18260 Glycine max

AN53

POPTR_0004s16280 Populus trichocarpa

POPTR_0005s22720 Populus trichocarpa

POPTR_0002s05810 Populus trichocarpa

AN42

AT3G05570 Arabidopsis thaliana

At2g21640 Arabidopsis thaliana

AT4G39235 Arabidopsis thaliana

VIT_03s0063g02630 Vitis vinifera

AN37

Solyc02g085260.1 Solanum lycopersicum

Solyc01g109680.2 Solanum lycopersicum

PHYPADRAFT_19838 Physcomitrella patens

Figure 3.2 B) UPOX1 orthologs. (A) Phylogenetic tree inferred from a simultaneous

comparison of UPOX1 orthologs based on sequence similarity. The tree was

constructed by PANTHER. PANTHER defines a controlled vocabulary for protein anno-

tation (the PANTHER/X ontology), as well as a method for classifying new sequences

(scoring against the PANTHER/LIB library of Hidden Markov Models).

Chapter 3 Expression and regulation of UPOX1

104

0

5

10

15

20

25

30

UPOX1

Rela

tive

tra

nsc

rip

ta

bu

nd

an

ce

* *

*

*

**

Time (hours)

0

1

2

3

4

5

6

7

8

903 12 24

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

5

0 3 12 24

0

3 12 240

UPOX2

UPOX3

* **

Figure 3.3 Transcript abundance of UPOX 1, UPOX 2 and UPOX 3.

QRT-PCR analysis of transcript abundance for UPOX1 homologous

genes over 24 h in response to the addition of citrate, H2O2, rotenone and

salicylic acid to Arabidopsis cell cultures. The amount of transcript prior

to the addition of compounds was set to 1 and changes expressed in a

relative manner. An asterisk indicates a significant difference

(P ≤ 0.05) relative to the 0 h timepoint for each individual treatment.

*

Citrate

H2O2

Rotenone

Salicylic Acid

Chapter 3 Expression and regulation of UPOX1

105

0

2

4

6

8

0 3 12 24

Citrate

H2O2

Rotenone

Salicylic Acid

0

2

4

6

8

10

0 3 12 24

0

510

15

20

2530

35

40

0 3 12 24

AOX1a NDB2

ATOM66

Rela

tiv

etr

an

sc

rip

tab

un

dan

ce

*

** *

* * *

**

*

* *

**

**

*

** *

*

*

* **

* *

*

* *

*

0

0.4

0.8

1.2

1.6

0 3 12 24

UBC

*

*

**

* * * **

*

*

0 3 12 240

20

40

60

80

100

120

140

0 3 12 24

PR1

Time (hours)

2.5

1.5

0.5

*

*

*

*

* *

*

**

*

*

Rela

tiv

etr

an

sc

rip

tab

un

dan

ce

Rela

tiv

etr

an

scri

pt

ab

un

dan

ce

Time (hours) Time (hours)

Time (hours)

Figure 3.4 Transcript abundance of AOX1a, NDB2, ATOM66, UBC and

PR1a. QRT-PCR analysis of transcript abundance for various genes over 24 h

in response to addition of citrate, H2O2, rotenone and salicylic acid in Arabidop-

sis cell cultures. The amount of transcript prior to the addition of compounds

was set to 1 and changes expressed in a relative manner. An asterisk indicates

a significant difference (P ≤ 0.05) relative to the 0 h timepoint for each individual

treatment and gene.

Time (hours)

Chapter 3 Expression and regulation of UPOX1

106

0

5

10

15

20

25

0h 1h 3h 6h

UPOX1

Aox1a

Rela

tive

tran

script

ab

und

an

ce

Figure 3.5 Transcript abundance of UPOX 1 and Aox1a. QRT-PCR

analysis of transcript abundance for UPOX1 and Aox1a in response to the

application of UV light stress at 280nm. An asterisk indicates a significant

difference relative to the 0 h timepoint for each gene, (P ≤ 0.05).

*

**

*

Time (h) after treatment

Chapter 3 Expression and regulation of UPOX1

107

Mutations

Figure 3.6 Relative transcript abundances of UPOX1, UPOX2, UPOX3, AOX1a,

NDB2, AtOM66, PR1 and UBC. The relative expression data for these genes as viewed

as a heat map in response to stimulus and mutations using Genevestigator. Displayed

are the top 50 (most upregulated and down regulated) stress treatments and mutations.

Red indicates an increase and green indicates a decrease in abundance.

Stimulus

UP

OX

1

UP

OX

2

UP

OX

3

AO

X1

a

ND

B2

AT

OM

66

PR

1

UB

C

UP

OX

1

UP

OX

2

UP

OX

3

AO

X1

a

ND

B2

AT

OM

66

PR

1

UB

C

Chapter 3 Expression and regulation of UPOX1

108

Figure 3.7 Expression of AOX1a, NDB2, UPOX, ATOM66, PR1a, and UBC in

defence signalling mutants. The relative transcript abundance was determined

by qRT-PCR in the defense signalling mutants etr1, abi3, pad4, npr1, jar1, and

eds1, and NahG and compared to Col-0 where expression was set to 100%.

Experiments conducted in triplicates.

ATOM66

UBC

PR1

UPOX1

NDB2

AOX1a

Chapter 3 Expression and regulation of UPOX1

109

Figure 3.8 Histochemical localisation of GUS activity in Arabidopsis

transformed with the full-length UPOX1 upstream region fused to the GUS

reporter gene. A) Inflorescence (flower stalk). B) Root tip. C) Emerging lateral

root primordium. D) Flower/pedicel. E) Seed Silique. F) Anthers at anthesis

A) B) C)

D) E) F)

Chapter 3 Expression and regulation of UPOX1

110

CT

TA

A

CA

AC

A

TTA

AG

GT

CA

AT

TG

AC

WR

KY

RA

V1

Str

ess

Resp

onsiv

e

ele

men

ts

AC

GT

GI

ele

men

t

CT

TA

A

AA

CA

AA

CM

YB

CA

GA

T

CG

TG

AT

Be

lem

ent

-400

AT

G-

20

0-

10

00

-8

00

CT

TA

AC

GT

GA

T

AC

GT

G

CT

TG

GC

CA

CG

-60

0

GT

CA

A

CT

TA

A

GT

CA

A

AA

CA

AA

C

CA

AC

A

CT

TA

A

CT

TA

A

CT

TA

A

CA

GA

T

UP

OX

1U

pstr

ea

mre

gio

n

Fig

ure

3.9

Su

mm

ary

of

the

pre

dic

ted

cis

-acti

ng

reg

ula

tory

ele

me

nts

inth

eU

PO

X1

pro

mo

ter

reg

ion

.P

uta

tive

cis

-acting

reg

ula

tory

ele

men

ts

were

identified

inth

eU

PO

X1

pro

mote

rre

gio

nu

sin

ga

va

rie

tyof

pre

dic

tion

meth

ods.

Nu

mberin

gis

fro

mth

etr

ansla

tio

na

lsta

rtsite.

The

ele

ments

pre

dic

ted

inclu

ded

the

MY

B,

WR

KY,

RA

V1

an

dstr

ess

resp

onsiv

ee

lem

ents

.E

lem

ents

Ban

dI

that

ha

ve

bee

np

revio

usly

iden

tified

tobe

active

in

the

AO

X1a

pro

mote

r,th

ese

ele

men

tsw

ere

als

oid

entified

tobe

inth

eU

PO

X1

pro

mo

ter.

Chapter 3 Expression and regulation of UPOX1

111

20

100

80

60

40

140

120

0

0

50

100

150

200

250

250

GU

SA

ctiv

ity

Mock

H2O2

Rotenone

**

*

*

*

*

*

***

0

50

100

150

200

250

Cell Culture

GU

SA

ctiv

ity

20

100

80

60

40

140

120

0

Leaves

Figure 3.10 Functional analysis of the B and I predicted motifs in the promoter of

UPOX1. Sequence elements identified in the promoter region of UPOX1 were tested for

functionality by deleting the elements and testing the ability of the mutated promoter to

drive GUS expression. Cell cultures and 4 week old Col-0 leaf tissue were used to for

biolistic transformation assays. GUS activity of the untreated wild-type promoter was

normalised to 100% with all other activities expressed as a percentage of this. A red

asterisk indicates a significant difference between the GUS activity of the wild-type

promoter and the mutated promoter within a treatment. A green asterisk indicates a

significant difference between the GUS activity of a mock-treated versus treated

samples within a promoter. Significance is calculated using the student's t test with a P

value ≤ 0.05.

35S

35S

**

*

*

*

*

*

*

UPOX1 wt

UPOX1I

UPOX1B

UPOX1I +

B

UPOX1 wt

UPOX1I

UPOX1B

UPOX1I +

B

Chapter 3 Expression and regulation of UPOX1

112

Nu

cle

us

H2O

2

Ro

t

AO

X1

a

ATO

M66

UP

OX

1

ND

B2

AO

X1

a

AtO

M66

UP

OX

1

ND

B2

Cyto

pla

sm

AA

AA

AA

AA

AA

AA

AA

AA

TS

S

TS

S

TS

S

TS

S+++

+

+-

+

ED

S1

PA

D4

S.A

Fig

ure

3.1

1A

su

mm

ary

of

the

sig

nallin

gp

ath

ways

that

reg

ula

testr

ess

ind

uced

tran

scri

pt

ab

un

dan

ce

for

UP

OX

1an

dco

-exp

ressed

gen

es

en

co

din

g

mit

och

on

dri

alp

rote

ins.T

hre

edis

tinctive

path

ways

regula

tem

itochondrials

tress

response

ata

transcriptionall

evel,

an

SA

-dependentpath

way

thatis

repre

-

sente

dby

AtO

M66.

The

second

path

way

repre

sents

aconverg

ence

poin

tfo

rsig

nals

genera

ted

by

H2O

2and

rote

none

that

act

via

the

sam

eC

AR

Es.

H2O

2

and

rote

none

act

at

atr

anscriptional

level.

There

are

diffe

rences

inth

isactions

betw

een

genes

nam

ely

rote

none

als

ohas

are

pre

ssiv

eeffect

on

specific

regio

ns

ofth

eA

OX

1a

pro

mote

r,how

ever

rote

none

has

no

directeffecton

the

pro

mote

rofU

PO

X.T

he

third

path

way

thatacts

via

ED

S1

and

PA

D4

and

regu-

late

sonly

AO

X1a.

The

AO

X1a

pro

mote

ris

str

ongly

repre

ssed

under

norm

alconditio

ns

and

isde-r

epre

ssed

follo

win

gtr

eatm

ent.

The

regula

tion

of

transcript

abundance

follo

win

gtr

eatm

ent

with

SA

isre

gula

ted

at

atr

anscriptionalle

velfo

rU

PO

X1,A

tOM

66

and

post

transcriptionally

for

AO

X1a

and

ND

B2.

SA

has

a

sm

all

butsig

nific

anteffecton

the

UP

OX

pro

mote

rand

this

may

be

indirectby

trig

gering

oxid

ative

str

ess.

Chapter 4 Protein Function and Localisation

113

Chapter 4

Protein Function and Localisation

Chapter 4 Protein Function and Localisation

114

4.1 Introduction

To determine the intra-organelle localisation of UPOX1 several in vivo and in vitro

methods were utilised, including GFP-localisation, import assays and BN-PAGE

analysis. To investigate this stress responsive gene at a functional level several

under-expressing lines and over-expressing lines were generated. To confirm these

lines, Western blots were performed with an antibody which was raised against

UPOX1, see Section 2.10.3. Furthermore, the UPOX1 over-expressing and

under-expressing lines were used in BN-PAGE gels in order to observe possible

proteins that may associate with UPOX1 in its native form.

The results from the in vitro assays were all visualised on either SDS-PAGE or

BN-PAGE gels. SDS-PAGE gels separate proteins according to their electrophoretic

mobility which is a function of the length of the polypeptide chain or molecular weight.

The detergent used is sodium dodecyl sulphate denatures proteins and provides a

negative charge to the protein that is proportional to its mass (Davis and Ornstein,

1959). Native PAGE separations are contrastingly run under non-denaturing

conditions with no detergents. Therefore complexes remain associated and folded as

they would otherwise in the cell. For BN-PAGE gels Coomassie Brilliant Blue dye is

used to provide a charge to the protein complexes during electrophoretic separation

(Schägger and von Jagow, 1991). Therefore, by performing all these assays it’s

possible to obtain a more thorough and in depth understanding of the intra-organelle

location of UPOX1.

Chapter 4 Protein Function and Localisation

115

4.2 Aims and Strategies

The specific aim of the research outlined in this chapter was to confirm and

characterise the function, location and interaction of UPOX1 within the plant cell.

This was conducted by performing;

- Generation and analysis UPOX1 T-DNA under-expressing lines, UPOX1

over-expressing line and Col-0 wild type

- In vivo GFP assays

- In vitro mitochondrial import assays

- In vitro chloroplast import assays

- Alkaline extractions

- Proteinase K titrations

- Blue Native PAGE assays to determine if UPOX1 is bound to a particular complex

Chapter 4 Protein Function and Localisation

116

4.3 Results

4.3.1 Generation of UPOX1 under-expressing

(knockdown) and over-expression lines

In order to characterise the function of UPOX1, multiple knock down-lines were

generated including an artificial microRNA (AmiRNA) line and an UPOX1

over-expressing line. Furthermore, several under-expressing lines were also sourced

from the Arabidopsis Biological Research Center (ABRC) including SALK_10761 and

SALK_125930. To determine if the site of the T-DNA insertions was within the exonic

coding region, the UPOX1 genomic regions were re-sequenced. DNA sequencing

confirmed that the T-DNA insertion was within the exonic region of UPOX1 for the

SALK_10761 line (Figure 4.1 A). The primers used are in Appendix 2. As the

SALK_125930 line did not contain its T-DNA insertion in the exonic regions it was not

used for further analysis. Homozygous stocks of the SALK_10761 line were grown in

soil mix pots according to Section 2.2.3, stratified for 3 days at 4ºC then grown under

long-day conditions (16 h light/8 h dark) at 22ºC. The plants were then confirmed to

be homozygous by PCR-based genotyping (primer sequences in Appendix 2), the

Sigma RedExtract PCR Kit Protocol and gel electrophoresis (according to the

methods outlined in Section 2.4.4) prior to being used for further transcriptomic and

proteomic analysis.

The amiRNA line was generated according to the methods outlined in Section 2.5 and

sequenced for confirmation. To determine the level of transcript downregulation

qRT-PCR analysis was conducted (Section 2.7). Following the qRT-PCR it was

concluded that the amiRNA line did not show a significant downregulation in the level

of UPOX1 transcript. Therefore, this line was not used for further analysis.

To examine the effect of increased levels of UPOX1 transcripts in plants, a

homozygous over-expressing line for UPOX1 was generated by Dr Olivier Van Aken

Chapter 4 Protein Function and Localisation

117

and Dr Inge De Clercq in the lab of Prof. Van Breusegem (VIB Ugent, Belgium) and

provided for use in this collaboration. This line was then consequently confirmed by

qRT-PCR for homozygosity prior to further analysis and use for this thesis (Section

2.7). Figure 4.1 B depicts the levels of UPOX1 in three lines including: the

SALK_10761 under-expressing line, the UPOX1 over-expressing line and Col-0 wild

type lines with UBC used as a housekeeping gene. From this qRT-PCR analysis it

was confirmed that the SALK_10761 under-expressing line showed a significant

decrease (approximately -90%) in UPOX1 transcript abundance relative to the Col-0

and the UPOX1 over-expressing line showed a significant increase (approximately

+400%) in UPOX1 transcript abundance relative to the Col-0.

The under-expressing, over-expressing and Col-0 lines were grown in water cultures

(according to the methods in Section 2.2.2) and purified mitochondria were purified as

described in Section 2.8.1. Mitochondrial proteins were separated on 16% SDS-PAGE

gels, transferred to PVDF membranes and probed with antibodies to UPOX1 and

Porin (generated by our laboratory). UPOX1 runs at an apparent molecular weight of

16 kDA on SDS-PAGE gels. Probing with the UPOX1 antibody resulted in a band that

was detected in mitochondria isolated from Col-0 plants but not from mitochondria

isolated from the UPOX1 under-expressing plants (Figure 4.2). A band with strong

intensity was detected in the over-expressing line with the same apparent molecular

mass as that detected in mitochondria isolated from Col-0 plants. Probing with the

Porin antibody control resulted in a cross-reacting band of uniform intensity in all three

mitochondrial samples. This position of this band corresponds to a molecular mass of

30 kDA. Thus the over-expressing and under-expressing lines were confirmed to

contain excess or no detectable quantities of UPOX1 protein respectively.

Under normal growth conditions (12 h light/12 h dark) at 22ºC, no phenotypic

differences existed between the Col-0 line, the under-expresser and over-expresser

lines as depicted in Figure 4.1 C. As UPOX1 is known to be a highly stress responsive

Chapter 4 Protein Function and Localisation

118

gene several stress assays were conducted under drought, salinity, heat and UV

exposure. To test these three lines under drought stress, plants were grown for 3

weeks and then water was withheld for 17 days following which the plants were re-

watered, this was then repeated. For salinity and heat stress phenotypic studies the

plants were treated according to the methods outlined in (Suter and Widmer, 2013)

and for UV stress 4 week old plants plants were treated with UV-C at 254 nm for 5

min. Additional stress treatments measuring root length and plant formation were also

conducted on MS-Agar plates according to the methods outlined in (Allu et al., 2014).

The left hand side agar plate shows the result of no UV-C stress and the agar plate on

the right hand side shows the effect of UV-C applied as mentioned above. There were

no phenotypic variations in either mutant line relative to the Col-0 line following these

stress treatments as can be seen in Figure 4.1 D or E.

4.3.2 In vivo GFP

In order to determine the organelle and intra-organelle localisation, chimeric proteins

were created by fusing GFP to the N and C-terminus of UPOX1 as described in

Section 2.9.1. These constructs were then biolistically transformed into Arabidopsis

suspension cell culture (Thirkettle-Watts et al., 2003; Lee and Whelan, 2004). As a

mitochondrial control, red fluorescent protein (RFP) was fused to the targeting signal

of soybean alternative oxidase (Aox-RFP) (Murcha et al., 2007) and the subsequent

fluorescence patterns were observed 48 h after transformation. The N terminal

UPOX1 GFP proteins displayed co-localisation with Aox-RFP following the merging of

their individual images, this indicated that UPOX1 was mitochondrially located (Figure

4.3A). Furthermore, the N-terminal UPOX1 GFP signal produced circular ring

structures (Figure 4.3 B), this can be indicative of proteins targeted to the outside of

the inner membrane or the outer mitochondrial membrane such as the outer

mitochondrially located protein Metaxin (Lister et al., 2007). N and C terminal GFP

Chapter 4 Protein Function and Localisation

119

constructs were also prepared and tested for UPOX2 and UPOX3. From the images

in Figure 4.3 it is clear that UPOX2 and UPOX3 are not localized to the mitochondria

in vivo as the two proteins do not overlap with the Aox-RFP control. Their appearance

is indicative of proteins localised to the cytosol.

4.3.3 In vitro mitochondrial imports

In vitro translation of the UPOX1 precursor protein generated a protein band on SDS-

PAGE gels with an apparent molecular mass of 16 kDa (Figure 4.4A, lane 1) with a

14 kDa band also present probably as a result of an alternative translation start. The

precursor protein was then incubated with freshly isolated mitochondria from 4 week

old Arabidopsis plants (lane 2). These conditions support in vitro import and the

protein was taken into the mitochondria. UPOX import is shown to be unaffected by

Valinomycin (Lane 4). Furthermore, upon rupture into mitoplasts (Lanes 6-9), UPOX

becomes exposed. This is evidence that the protein is not moving across the inner

membrane. This in vivo import is in correlation with the GFP results and suggests that

UPOX1 is not imported across the IMM and is localised to the OMM or IMS. As a

control, the TIM23-2 precursor protein was translated to generate a product with an

apparent molecular mass of 20 kDa. Also present was a 16kDa band that is a likely a

more protease resistant fragment of imported 23-2 (Figure 4.4B, lane 3). The addition

of valinomycin abolished import as evidenced by the fact that no protein was

protected by the addition of PK after the import assay (Figure 4.4B, lane 5). The outer

membrane was ruptured to confirm the protease protected protein was inserted into

the inner membrane. Following the rupture of the outer membrane and the addition of

PK a protease protected band with an apparent molecular mass of 16 kDa was

observed. This represented the portion of Tim23-2 that was present in the inner

membrane, protected from PK digestion. The addition of valinomycin to the import

Chapter 4 Protein Function and Localisation

120

assay followed by PK digestion of outer membrane ruptured mitochondria resulted in

no import being detected.

The import of AOX was also used a control. The AOX precursor protein was

translated to generate a product with an apparent molecular mass of 36 kDa. The

protein was imported and processed to produce a mature product of 32 kDa. The

mature protein was transported into the inner mitochondrial membrane as it was

confirmed to be PK resistant and sensitive to the addition of valinomycin (Figure

4.4C).

4.3.4 In vitro chloroplast import

In order to exclude the possibility that UPOX1 was also targeted to chloroplasts, i.e. a

dual targeted protein, in vitro import assays into chloroplasts were performed (Figure

4.5). The extensively studied small subunit of 1,5-ribulose bisphosphate

carboxylase/oxygenase (Rubisco SSU) was used as a control (Carrie et al., 2008).

Lane 1 for both UPOX1 and SSU show a precursor at 16 and 20 kDa respectively.

Following the incubation of the SSU precursor with chloroplasts a band was present at

20 kDa and a mature band was also present at 14 kDa. Following the addition of

thermolysin the mature band was present at 14 kDa showing that SSU was imported

into the chloroplast. Following the incubation of UPOX1 with chloroplast there appears

to be no band evident. Therefore it can be deduced from these experiments that

UPOX1 is not imported into chloroplasts and appears to be specifically imported into

the mitochondria.

4.3.5 Alkaline extractions

To determine if UPOX1 was located or associated in/with the mitochondrial

membrane or soluble fraction, alkaline extractions were performed according to

Section 2.9.5 (Figure 4.6). AOX was used as a control as it has been well established

that it is an inner membrane protein and therefore would be found in the insoluble

Chapter 4 Protein Function and Localisation

121

fraction (Lister et al., 2004). In vitro import of AOX and UPOX1 followed by alkaline

extraction resulted in AOX only being detected in the insoluble (membrane) fraction

while UPOX1 was detected in both the soluble and insoluble fractions (Figure 4.6).

This suggests that UPOX1 is associated with the outer membrane and aqueous

compartments or inter-membrane space of mitochondria.

4.3.6 Proteinase K titrations into mitochondria and

mitoplasts

In the previous import experiments, UPOX1 was partially resistant to PK treatment.

(As in Figure 4.4). In order to determine the efficiency of protease treatment, PK

titrations were carried out on mitochondria and mitoplasts. These were then immune

detected with several antibodies. The controls used included TOM20-2 which has

been previously localised to the outer mitochondrial membrane (Murcha et al., 1999;

Werhahn et al., 2001) AOX which has been previously localised to the inner

membrane and HSP 60 which is a matrix control protein (Millar et al., 2001) (Figure

4.7). Titrations ranging from 0 to 256 µg of PK were carried out per sample.

As expected, following PK titration the TOM20-2 protein depletes immediately; this is

to be expected as this protein is an outer mitochondrial protein and is immediately

sensitive to proteinase digestion. Protein bands are present for AOX and HSP60

following PK titrations on mitochondria and mitoplasts up to 128 µg of PK as expected

as they are located in the inner membrane and the matrix respectively. The amount of

UPOX1 diminishes with an increasing concentration of PK in mitochondria. Following

the rupture of the outer membrane of the mitochondria and the addition of PK no

protein bands are detectable. This suggests that UPOX1 may be located in or near to

the inner mitochondrial space. These results correlate with the in vitro import results

and the GFP analysis.

4.3.7 Blue Native PAGE

Chapter 4 Protein Function and Localisation

122

Blue Native Polyacrylamide gel electrophoresis (BN-PAGE) was conducted to

determine if UPOX1 was associated with a mitochondrial respiratory chain complex.

This experiment was conducted by performing BN-PAGE according to Section 2.11.2

BN PAGE sample preparation. BN-PAGE gels allow for the visualisation of the

mitochondrial multi-subunit respiratory chain complexes as they are prevented from

dissociation as no SDS is used (Swamy et al., 2006). The first panel shows the

Coomassie stained gel directly proceeding electrophoresis. The major respiratory

chain complexes and super complexes (CI+CIII, CI, CV, CIII, CIV and CII) are clearly

visible in Figure 4.8. For this analysis, mitochondria were isolated from Arabidopsis

Col-0 plants, the SALK_10761 UPOX1 under-expressing line and the UPOX1

over-expressing line.

These gels were then transferred onto PVDF membranes according to Section 2.11.3

Wet Transfer of BN PAGE gels and immune detected using several different

antibodies. For a complex I control NAD9 was used. BN-PAGE imports were carried

out to determine if UPOX1 was imported into any complexes. These gels were then

dried and exposed on Fuji BAS TR2040 plates. It was determined from these

experiments that UPOX1 was not imported into either complex I or the super complex

CI+CIII. Therefore the bands present for the UPOX1 western following BN-PAGE can

only be attributed to either minor associations without import or non-specific binding of

the antibody to these complexes. The import of UPOX1 and consequent BN-PAGE

for this result shows that there is a strong band very low in the BN-PAGE image. This

suggests that UPOX1 runs further into the gel than all the larger respiratory chain

complexes (without incorporation into the complexes).

4.4 Discussion

A number of approaches were undertaken to determine the sub-cellular localisation of

UPOX1. The mitochondrial localisation was confirmed using a number of methods,

Chapter 4 Protein Function and Localisation

123

namely western blot analysis, in vivo GFP tagging and in vitro uptake assays. The

intra-organellar localisation was investigated using alkaline extraction, PK digestions

and BN-PAGE. Several approaches are necessary to determine the location of

uncharacterised proteins as each of these techniques alone has its limitations and no

technique should be used alone. Only after several different approaches are taken

can a hypothesis be made about the organelle and intra-organelle location of any

protein of interest. In this case the results of all these approaches together correlate

with each other and suggests that UPOX1 is located in the mitochondrial

intermembrane space and may also be associated with the outer mitochondrial

membrane. Following phenotypic studies on the under-expressing and

over-expressing mutant lines in response to stress treatments no phenotypes were

detected. The lack of a phenotype could be explained by the compensation of

duplication genes (Gu et al., 2003) or by the mechanisms that allow stress networks

to buffer the effects of perturbations in related pathways and thereby allowing for

homeostasis to be maintained (Cutler and McCourt, 2005).

Chapter 4 Protein Function and Localisation

124

5‘U

TR

SA

LK

01

07

61

EX

ON

EX

ON

3‘U

TR

527

736

891

995

T-D

NA

Pri

mer

Set

874bp

02468

10

12

14

16

18

UP

OX

1U

BC

UP

OX

1T

-DN

AU

ndere

xpre

sser

SA

LK

_10761

Co

l-0

UP

OX

1O

vere

xpre

sser

RelativeTranscriptAbundance

40

*

*

A)

B)

UP

OX

1

Un

de

re

xp

resse

r

UP

OX

1

ove

re

xp

resse

rC

ol-0

UP

OX

1

Un

de

re

xp

resse

r

UP

OX

1

ove

re

xp

resse

rC

ol

-0

Co-0 U

POX1

overexp

ress

er

UPO

X1

Underexp

ress

er

Co-0

UPO

X1

overexp

ress

er

UPO

X1

Underexp

ress

er

E)

D)

C)

UV

No

UV

Fig

ure

4.1

Gen

era

tio

no

fu

nd

ere

xp

res

se

ran

do

vere

xp

ress

er

tran

sg

en

icli

nes

of

UP

OX

1.A

)T

he

ge

ne

str

uctu

reof

UP

OX

1,

the

T-D

NA

insert

is

with

inth

ee

xon

ofU

PO

X1

ge

ne

for

the

SA

LK

_1

07

61

un

de

rexp

resse

rlin

e.B

)q

RT

-PC

Ra

na

lysis

ofth

ere

lative

mR

NA

ab

und

an

ce

ofU

PO

X1

with

inth

e

T-D

NA

un

dere

xp

resser,

Col-

0an

dovere

xp

resser.

C)

Ph

eno

typ

iccom

parison

of4

we

ek

old

UP

OX

1o

ve

rexpre

sser,

Col-0

and

UP

OX

1u

nde

rexpre

sse

r

un

der

norm

alcon

ditio

ns.D

)C

)P

hen

oty

pic

com

pa

rison

of4

wee

ko

ldU

PO

X1

ove

rexpre

sser,

Co

l-0

an

dU

PO

X1

und

ere

xp

resser

un

de

r5

min

so

fU

V-C

at

254

nm

.P

hoto

gra

phe

d4

8h

after

tre

atm

ent.

E)

Phe

no

typic

com

pa

rison

an

dro

ot

gro

wth

of

the

UP

OX

1o

ve

re

xp

resse

r,C

o-0

and

UP

OX

1u

nde

r

exp

resse

r.P

lants

were

gro

wn

for

2w

ee

ks

vert

ically

und

er

no

trea

tme

nt

(Left

pla

te)

and

trea

ted

with

UV

-Cfo

r5

min

sa

t2

54

nm

(Rig

ht

pla

te).

Pho

to-

gra

phe

d48

hr

afte

rtr

ea

tmen

t.A

naste

risk

indic

ate

sa

sig

nific

antd

iffe

rence.

1

qR

T-P

CR

pri

mer

s

Chapter 4 Protein Function and Localisation

125

16 kDa

Col-0UPOX1 over expresser

30 kDa

Figure 4.2 Western blot analysis of isolated mitochondria from 2 week

old water culture plants confirming the mutant under expressing and

over expressing lines. The mitchondria were isolcated from Col-0 water

cultures, UPOX1 under expressing line SALK_10761 water cultures and the

UPOX1 over expressing line water cultures. Mitochondrial proteins were

probed with an antibody raised against UPOX1, Porin was used as a control.

UPOX1 under expresser

Chapter 4 Protein Function and Localisation

126

A)

Chapter 4 Protein Function and Localisation

127

B)

Figure 4.3 B) In vivo sub cellular localisation of N’ UPOX1. GFP fusion

construct using full length coding sequences were transformed into Arabidopsis

cultured cells using biolistic transformation. This image is a zoomed up image

from Figue 4.3 A) showing the circular ring structures.

Ring Structures

UPOX1GFP

10µm

Chapter 4 Protein Function and Localisation

128

Mito - + + + + + + + +

PK - - + - + - + - +

Val - - - + + - - + +

MP - - - - - + + + +

Lane 1 2 3 4 5 6 7 8 9

UPOX1

TIM23-2

AOX

16 kDa

36 kDa

16 kDa

32 kDa

20 kDa

Figure 4.4 Import of radiolabelled UPOX1, TIM23-2 and AOX into mitochon-

dria. Lane 1, precursor protein only. Lane 2, precursor protein incubated with

mitochondria under conditions that support import. Lane 3, as lane 2 with 0.4

ug/ul proteinase K (PK) added. Lane 4, as lane 2 with 1 uM valinomycin (Val)

added. Lane 5, as lane 2 with PK and Val added. Lane 6 to 9 as lanes 2 to 5

respectively with ruptured outer mitochondrial membranes.

14 kDa

A)

B)

C)

Chapter 4 Protein Function and Localisation

129

Chapter 4 Protein Function and Localisation

130

AOX

Lane

UPOX1

1 2 3

Mitochondria +_ +

Figure 4.6 Alkaline extractions of AOX and UPOX1. Alkaline

extractions following in vitro imports assays. Lane 1, precursor

protein alone. Lane 2, soluble fraction. Lane 3, insoluble fraction

Chapter 4 Protein Function and Localisation

131

Tom20-2

AOX

HSP 60

0 16 32 64 128 256

Lane

PK 0 16 32 64 128

1 2 3 4 5 6 7 8 9 10 11

MP - - - - - - + + + + +

Figure 4.7 Western blot analysis of proteinase K titrations of TOM20-2, AOX, HSP

60 and UPOX1. Both mitochondria and mitoplast (MP) samples were treated with

increasing amounts of PK. Lane 1, mitochondria with no PK added, Lanes 2, 3, 4, 5

and 6 as lane 1 with the addition of 16, 32, 64, 128 and 256ug PK respectively. The outer

membrane was ruptured via osmotic shock for lanes 7 to 12. Lane 7 as lane 1 except

that mitochondria were osmotically shocked prior to PK digestion. Lanes 8, 9, 10 and 11

as lanes 2-5 except that mitochondria were osmotically shocked prior to PK digestion.

UPOX1

23 kDA

30 kDa

60 kDA

16 kDA

Chapter 4 Protein Function and Localisation

132

CV

CII

I

CI+

CII

I

CIVC

I

UPOX1KOCol-O

UPOX1KOCol-O

UPOX1ovx

UPX1ovx

UPOX1KO

Col-O

UPOX1ovx

Co

om

assie

sta

ine

dg

el

UP

OX

1

An

tib

od

y

NA

D9

An

tib

od

y

UPOX1KO

Col-O

UPOX1ovx

Pro

hib

itin

An

tib

od

y

UPOX1KO

Col-O

UPOX1ovx

UP

OX

1

Imp

ort

UPOX1KO

Col-O

UPOX1ovx

Pro

hib

itin

Impo

rt

CII

Fig

ure

4.8

Blu

eN

ati

ve

PA

GE

ex

am

ina

tio

no

fC

ol-

0(w

ild

typ

e),

UP

OX

1S

AL

K_

10

761

T-D

NA

kn

ock

ou

ta

nd

UP

OX

1o

ve

r

exp

ressin

gA

rab

ido

ps

islin

es

.T

he

firs

tp

ane

lshow

sa

co

om

assie

sta

ino

fa

BN

-PA

GE

gel,

pan

els

2to

4d

ep

ict

BN

-PA

GE

ge

ls

follo

win

ga

weste

rnusin

gth

eU

PO

X1

,N

AD

9an

dP

roh

ibitin

antibo

die

sre

spe

ctively

.P

anels

5an

d6

sh

ow

the

pro

duct

of

BN

-PA

GE

ge

lsfo

llow

ing

imp

ort

su

sin

gU

PO

X1

an

dP

roh

ibitin

pre

cu

rso

rsre

sp

ective

ly.

21

34

56

Chapter 5 General Discussion

133

Chapter 5

General Discussion

Chapter 5 General Discussion

134

5.1 Summary of Aims and Approaches

This study involved carrying out the most comprehensive and in-depth research on

UPOX1 to date. The aims of this project involved taking a multi-disciplinary approach

in order to characterise UPOX1, this has incorporated various transcriptomic,

proteomic and bioinformatic approaches. Specifically, the approaches taken included:

1) Investigations into the kinetics of UPOX1 induction with the aim of identifying

pathways that regulate stress induced gene expression. These assays were

compared and contrasted against other known stress responsive genes such

as AOX1a and other mitochondrial proteins such as external NAD(P)H-

dehydrogenase NDB2 and AtOM66.

2) Transcript analysis was conducted on defense signalling mutants to determine

the pathways that regulates this stress induced gene.

3) The promoter regions were examined to determine the functional CAREs that

play an active role in the response to stress treatments such as with H2O2,

rotenone, salicylic acid and combinations of these.

4) The co-expression and co-regulation of UPOX1 was examined.

5) Several proteomic methods were employed to determine the localisation of

UPOX1 within the plant cell and more specifically the mitochondria. These

approaches involved; conducting in vivo GFP analysis in conjunction with

antibody based in vitro isolation methods

6) The use of an over-expressing line and under-expressing line for UPOX1 for

functional analysis.

5.2 Summary of Results

UPOX1 to date in scientific journals is still often categorised as an ‘unknown stress

responsive gene’. The research conducted in this PhD has added valuable, new and

Chapter 5 General Discussion

135

in depth knowledge on this gene, its corresponding protein, how it is co-regulated and

where it is located within the cell.

Specifically it has been elucidated that the homologs of UPOX1, (UPOX2 and

UPOX3) differ considerably from the qRT-PCR transcript profiles of UPOX1.This

suggests that the regulatory mechanisms controlling the expression of each gene may

be largely unrelated and/or independent. In addition, qRT-PCR assays also showed

that UPOX1 was found to be significantly more stress responsive than its two

homologs. Further investigations focused on the kinetics of transcript induction and it

was established that UPOX1 following the treatment of a wide array of stress

treatments responded late as it peaked 24 h after stress treatment. From these

studies, it was concluded that UPOX1 was involved in a secondary stress response

pathway. The kinetics of induction were also tested for several other putatively co-

expressed genes and following these qRT-PCR assays it was confirmed that the

pattern of transcript induction for NDB2 and AOX1a differed from that of UPOX1.

Investigations were carried out to determine if UPOX1 was co-expressed or

co-regulated with any other genes. It was first confirmed that of all the genes tested

AOX1a was the gene that was most co-expressed with UPOX1. This suggested that

there is a possible overlap in the pathways responsible for the induction of these

genes. Following on from this, research was carried out to confirm whether UPOX1

and AOX1a were co-regulated. These experiments involved initially narrowing down

putative functional elements in the upstream regions and mutating these regions.

Biolistic transformation assays confirmed that UPOX1 and AOX1a share the same

functional motif in their upstream region and therefore these genes were confirmed as

being co-regulated.

In compiling the transcriptomic and biolistic transformation data three pathways that

mediate mitochondrial stress response were identified. The first was a SA dependent

pathway in which only AtOM66 is regulated at a transcriptional level while UPOX1,

Chapter 5 General Discussion

136

AOX1a and NDB2 are regulated at a post-transcriptional level. The second pathway is

the lack of response by UPOX1 to rotenone which differs significantly to AOX1a. The

third pathway is represented by AOX1a as its expression differs from UPOX1 and

NDB2, this suggests a different pathway involved via EDS1 and PAD4 to regulate only

AOX1a. From these investigations into the promoter elements in UPOX1, AOX1a and

NDB2 it was concluded that SA acts to increase transcript abundance of these via a

post-transcriptional mechanism in contrast to AtOM66 which is under direct

transcriptional control. Finally it was also found that H2O2 and rotenone can both act

transcriptionally and their CAREs overlap however different transcription factors are

thought to be involved for UPOX1, AOX1a and NDB2. Therefore transcript analysis,

defense signally mutants and promoter activity studies together have provided new

and detailed insights into the intricate mechanisms underlying the expression of these

genes under stress conditions.

By conducting western blot analysis, in vivo analysis, GFP tagging, in vitro uptake

assays along with alkaline extractions, PK digestions and BN-PAGE it was confirmed

that UPOX1 is located in the mitochondrial intermembrane space and may also be

associated with the outer mitochondrial membrane. Interestingly, UPOX1 seems to be

associated with both membrane and soluble fractions. It has been recently found by

ID Blue Native PAGE that UPOX1 may also be associated with the membrane arm of

Complex I (Senkler et al., 2016).

5.3 Functional implications of the

mitochondrial stress response

Mitochondria have several crucial roles in the functioning of plants; these include the

integration of carbon and nitrogen into the metabolism of plants, the biosynthesis of

pyrimidines, heme, vitamins, iron-sulphur centres. Additionally they are involved in

Chapter 5 General Discussion

137

signalling programmed cell death and the production of ATP during phosphorylation

and the TCA cycle. Several studies over the years have demonstrated how

mitochondria are the site of and target of a variety of stresses (Bartoli et al., 2004;

Giraud et al., 2008) yet there is still very little known about many of the smaller

proteins that are encoded in the nucleus, targeted to the mitochondria and are stress

responsive. This thesis has focused on one such protein UPOX1, a hallmark of

oxidative stress (Gadjev et al., 2006) and using a multitude of approaches many of the

mitochondrial stress pathways in which it is involved in have been deduced. A main

sub-topic in relation to mitochondrial stress response (MSR) has revolved around the

production of ROS species and the targeting of ROS as it is known to critically

damage proteins, nucleic acids and lipids (Fujita et al., 2006). Within our lab it has

been shown that the activity of AOX in conjunction with NAD(P)H dehydrogenases

enable the ETC to continue functioning by preventing over reduction of the

constituents of the ETC, furthermore, it has also been shown that the lack of AOX

produces a heightened sensitivity to combined drought and light stress in Arabidopsis

(Giraud et al., 2008; Van Aken et al., 2009).

AOX1a, the alternative respiratory pathway and the ETC have been studied

extensively in relation to MSR however it is now clear that MSR pathways are much

wider than just encompassing these components. For example the mitochondrial

substrate carrier protein family (MCF) are the strongest represented protein family in

MSR pathways. The nature of the MCFs by their broad stress responsiveness which

may be involved in Pi (At3g48850), ADP/ATP (At4g28390) and dicarboxylic acid

transport suggests that under stress conditions, mitochondria requires an increased

exchange of respiration and TCA cycle substrate (At4g27940 and At2g22500) (Hamel

et al., 2004; Palmieri et al., 2008).

In order to gain a further understanding of the functional implications of the

mitochondrial stress response 26 mitochondrial stress responsive genes, including

Chapter 5 General Discussion

138

UPOX1 were investigated within the Whelan lab and these genes were examined in

16 selected microarray datasets (Van Aken et al., 2009). The 16 selected microarrays

spanned various stress conditions including heat, UV, ozone, salt and osmotic stress.

Cluster analysis was conducted on this data as information about a gene’s function

can be deduced by identifying genes that share its expression pattern. Genes that are

co-expressed under a variety of different circumstances may work together in the cell,

they may be involved in a co-ordinated activity and may encode proteins that are part

of the same multi-protein machine. The cluster analysis showed that in all conditions

tested there was a predictable trend of up regulation for most genes although a few

treatments that are often considered to be physiologically comparable such as salt

and osmotic stress did not show a similar transcriptomic response. For example

UPOX1 and sHSP23.5 were observed to be strongly down regulated during osmotic

stress although up regulated during salt stress. Saying this however, most other cases

did show comparable treatments clustering together (Van Aken et al., 2009).

In terms of gene specific expression patterns, the clustering analysis revealed

correlations that reveal functional implications for UPOX1 and mitochondrial import

pathways. UPOX1 and Tim17-1 were shown to be highly co-expressed. Tim17-1 is

the only import component known to date to be altered by stress. Another interesting

note to mention is that UPOX1 along with most other mitochondrial proteins are

imported into the mitochondria and therefore it’s likely that the rate of mitochondrial

import changes during stress.

The clustering analysis also revealed that UPOX1 is also co-expressed with

sHSP23.6, sHSP23.5, AOX1a and NDB2 (Van Aken et al., 2009). As members of the

alternative respiratory pathway AOX1a and NDB2 can form a complete respiratory

chain, dissipating energy as heat whilst not contributing to ATP synthesis. The

co-expression of AOX1a, NDB2 with UPOX1 has been documented before in (Clifton

et al 2005) and the research conducted in this thesis shows the extent to which

Chapter 5 General Discussion

139

UPOX1 and AOX1a are also co-regulated as a result of CAREs within their promoter

regions. This implies that the observed expression patterns of MSR genes are at least

partially regulated by underlying co-regulation mechanism (Van Aken et al., 2009).

Additional studies have investigated the expression of known stress responsive genes

in mutant atphb3 lines on (Van Aken et al., 2010). Both AOX1a, a marker of

mitochondrial retrograde signalling in plants as well as UPOX1 were significantly

upregulated (UPOX1 was upregulated 40-fold). This further indicates that at least part

of the mitochondrial stress response can be triggered by retrograde signalling.

5.4 Retrograde regulation of nuclear genes

encoding mitochondrial proteins during

stress and the mitochondrial dysfunction

stimulon

Many molecular components of retrograde signalling pathways that regulate

expression of nuclear genes encoding mitochondrial proteins (NGEMP) are known to

interact with chloroplast, growth and stress signalling pathways in the cell at different

levels. Some components are involved in the transmission and execution of these

signals. This positions mitochondria as important hubs for signalling in the cell, not

only in direct signalling of mitochondrial function but also in sensing and/or integrating

a variety of other internal and external signals. This integrates growth with energy

metabolism and stress responses.

A review by Ng et al., 2014 analysed the expression characteristics of all NGEMPs in

response to chemical and genetic mitochondrial perturbations. The MRR signalling

pathways were assessed and from this meta-analysis at least two transcriptomic

footprints were apparent from the clustering: genes that are generally up-regulated

and genes that are generally down-regulated under most of the conditions. As

Chapter 5 General Discussion

140

predicted UPOX1 was upregulated over all five categories, including abiotic, biotic,

nutrient, chloroplast and mitochondrial perturbations. More specifically this included

UV, salt, water and oxygen availability, temperature and many pathogens (Ng et al.,

2014).

Some of these genes identified as being generally stress responsive were also part of

the so-called Mitochondrial Dysfunction Stimulon (MDS) (De Clercq et al., 2013) of

which UPOX1 is a member along with MGE1 (At5g55200), sHsp23.5 and AtOM66

(Van Aken et al., 2009). The genes belonging to the MDS share a common cis-

regulatory element in their promoters that have been named the mitochondrial

dysfunction motif (MDM). This arbitrates the MRR-mediated induction by NAM, ATAF

½ and CUC2 (NAC) transcription factors (De Clercq et al., 2013; Ng et al., 2014). Of

these, the transcription factors ANAC017, ANAC013, ANAC053 and ANAC078 both

belonging to the transcription factor class NAM target UPOX1 (De Clercq et al., 2013).

It has been suggested that there is an integration of multiple MRR pathways at the

transcriptional or more upstream level as the MDM seems to be sufficient for

responsiveness to various genetic mitochondrial perturbations such as the prohibitin

mutant atphb3 (Van Aken et al., 2007; Van Aken et al., 2016), pharmacological

perturbations such as rotenone, Antimycin A and H2O2. Furthermore, a W-box cis-

element is also present in the MDS promoters and the W-box binding WRKY

transcription factors regulate the expression of these genes under various conditions

including many mitochondrial perturbations and high-light (Van Aken and Whelan,

2012; Van Aken et al., 2013).

Further investigations within the lab have suggested that the MDS genes act as a

non-specific and primary defense layer in response to a multitude of stress conditions

by avoiding (through alternative respiration) and by alleviating the effects of oxidative

stress, which is a common factor during different adverse conditions (Giraud and Van

Aken, 2012). Genome wide transcription studies by Rhoads and Subbaiah, 2007;

Chapter 5 General Discussion

141

Schwarzlander et al., 2012 also concur with the hypothesis that multiple MRR

pathways are present. This hypothesis has been further re-enforced by the promoter

analysis in this thesis that have tested transcript abundances of four stress responsive

NGEMPs (AOX1a, AtOM66, NDB2, and UPOX1) in response to rotenone, H2O2, and

salicylic acid and have found that discrete regulatory mechanisms exist.

In the last few years more details on how transcription factors control their target

genes have been elucidated. For example; AtWRKY40 has been shown to limit the

induction of UPOX1, AOX1a, NDB2 and AtOM66 after high light and AA stress

treatments and AtWRKY63 has been shown to positively affect the expression of

these genes (Van Aken et al., 2013). Furthermore, the expression of UPOX1 along

with AOX1a and several other genes encoding mitochondrial and non-mitochondrial

proteins have been found to be positively regulated by a group of NAC transcription

factors. These NAC transcription factors are bound to the endoplasmic reticulum

membrane and are released following mitochondrial inhibition. This discovery helps to

explain how signals could be transmitted from mitochondria to the nucleus as the

location of the ER bound NAC transcription factors associated with actin filaments

facilitates a close interaction between mitochondria and the ER (Ng et al., 2014).

5.5 Future Directions

The understanding of the molecular components that control the expression of

NGEMPS’s has significantly progressed in the last decade. This thesis continues to

add to the knowledge base but there is much we still don’t understand as only a

portion of these regulators and their gene targets are known to date. Several more

studies incorporating various stress responsive genes, transgenic lines and promoter

analysis experiments are required to demonstration a comprehensive understanding

of this field.

Chapter 5 General Discussion

142

In summary, the data presented in this thesis comprises the most comprehensive,

integrated analysis of the highly stress responsive hallmark of oxidative stress UPOX1

performed to date. In this study, this previously un-characterised protein was found to

be located in the mitochondria, elucidated the transcript kinetics and identified various

pathways in which it is involved. To investigate and characterise the function of

UPOX1 further advanced proteomic and metabolomic approaches could be used in

conjunction with stress treatments and new transgenic lines. Other future approaches

could involve the identification of potential protein interaction partners of UPOX1 and

genome-wide expression studies of the UPOX loss- and gain-of-function lines under

specific stress conditions.

Chapter 5 General Discussion

143

References

Adams K, Qiu Y, Stoutemyer M, Palmer J (2002) Punctuated evolution of mitochondrial gene content: high and variable rates of mitochondrial gene loss and transfer to the nucleus during angiosperm evolution. Proceedings of the National Academy of Sciences of the United States of America 99: 9905

AGI (2000) Analysis of the genome sequence of the flowering plant Arabidopsis thaliana. Nature 408: 796-815

Aitchison J, Nuttley W, Szilard R, Brade A, Glover J, Rachubinski R (1992) Peroxisome biogenesis in yeast. Molecular microbiology 6: 3455-3460

Allu AD, Soja AM, Wu A, Szymanski J, Balazadeh S (2014) Salt stress and senescence: identification of cross-talk regulatory components. Journal of experimental botany: eru173

Bahar O, Mordukhovich G, Luu DD, Schwessinger B, Daudi A, Jehle AK, Felix G, Ronald PC (2016) Bacterial outer membrane vesicles induce plant immune responses. Molecular Plant-Microbe Interactions 29: 374-384

Balmer Y, Koller A, Val G, Schürmann P, Buchanan BB (2004) Proteomics uncovers proteins interacting electrostatically with thioredoxin in chloroplasts. Photosynthesis Research 79: 275-280

Barah P, Jayavelu ND, Sowdhamini R, Shameer K, Bones AM (2015) Transcriptional regulatory networks in Arabidopsis thaliana during single and combined stresses. Nucleic acids research: gkv1463

Bartels D, Dinakar C (2013) Balancing salinity stress responses in halophytes and non-halophytes: a comparison between Thellungiella and Arabidopsis thaliana. Functional Plant Biology 40: 819-831

Bartoli CG, Gomez F, Martinez DE, Guiamet JJ (2004) Mitochondria are the main target for oxidative damage in leaves of wheat (Triticum aestivum L.). Journal of experimental botany 55: 1663

Bauer NC, Doetsch PW, Corbett AH (2015) Mechanisms Regulating Protein Localization. Traffic

Beal MF (2005) Mitochondria take center stage in aging and neurodegeneration. Annals of neurology 58: 495-505

Belt K, Huang S, Thatcher LF, Casarotto H, Singh K, Van Aken O, Millar AH (2017) Salicylic acid-dependent plant stress signalling via mitochondrial succinate dehydrogenase. Plant Physiology: pp. 00060.02017

Ben-Shachar D, Laifenfeld D (2004) Mitochondria, synaptic plasticity, and schizophrenia. International review of neurobiology 59: 273-296

Benikhlef L, L’Haridon F, Abou-Mansour E, Serrano M, Binda M, Costa A, Lehmann S, Métraux J-P (2013) Perception of soft mechanical stress in Arabidopsis leaves activates disease resistance. BMC plant biology 13: 1

Bergthorsson U, Adams K, Thomason B, Palmer J (2003) Widespread horizontal transfer of mitochondrial genes in flowering plants. Nature 424: 197-201

Berná G, Robles P, Micol JL (1999) A mutational analysis of leaf morphogenesis in Arabidopsis thaliana. Genetics 152: 729-742

Berthold DA, Andersson ME, Nordlund P (2000) New insight into the structure and function of the alternative oxidase. Biochimica et Biophysica Acta (BBA)-Bioenergetics 1460: 241-254

Bostock RM (2005) Signal crosstalk and induced resistance: straddling the line between cost and benefit. Phytopathology 43

Bowling SA, Clarke JD, Liu Y, Klessig DF, Dong X (1997) The cpr5 mutant of Arabidopsis expresses both NPR1-dependent and NPR1-independent resistance. The Plant Cell Online 9: 1573

Chapter 5 General Discussion

144

Bowsher CG, Tobin AK (2001) Compartmentation of metabolism within mitochondria and plastids. Journal of experimental botany 52: 513

Bruce BD, Perry S, Froehlich J, Keegstra K (1994) In vitro import of proteins into chloroplasts. Plant molecular biology manual 1: 1–15

Brunk C (1981) Mitochondrial proliferation during myogenesis* 1. Experimental Cell Research 136: 305-309

Butow RA, Avadhani NG (2004) Mitochondrial Signaling:: The Retrograde Response. Molecular cell 14: 1-15

Cannon W, Webb-Robertson BJO (2006) Computational Proteomics: High-Throughput Analysis for Systems Biology. In. World Scientific Pub Co Inc, p 403

Cao H, Bowling SA, Gordon AS, Dong X (1994) Characterization of an Arabidopsis mutant that is nonresponsive to inducers of systemic acquired resistance. The Plant Cell Online 6: 1583

Carrari F, Urbanczyk-Wochniak E, Willmitzer L, Fernie AR (2003) Engineering central metabolism in crop species::: learning the system. Metabolic Engineering 5: 191-200

Carrie C, Giraud E, Whelan J (2009) Protein transport in organelles: Dual targeting of proteins to mitochondria and chloroplasts. FEBS Journal 276: 1187-1195

Carrie C, Kühn K, Murcha MW, Duncan O, Small ID, O’Toole N, Whelan J (2009) Approaches to defining dual targeted proteins in Arabidopsis. The Plant Journal 57: 1128-1139

Carrie C, Murcha MW, Kuehn K, Duncan O, Barthet M, Smith PM, Eubel H, Meyer E, Day DA, Millar AH (2008) Type II NAD (P) H dehydrogenases are targeted to mitochondria and chloroplasts or peroxisomes in Arabidopsis thaliana. FEBS letters 582: 3073-3079

Chang C, Kwok SF, Bleecker AB, Meyerowitz EM (1993) Arabidopsis ethylene-response gene ETR1: similarity of product to two-component regulators. Science 262: 539

Chen S, Harmon AC (2006) Advances in plant proteomics. Proteomics 6: 5504-5516 Chen W, Provart NJ, Glazebrook J, Katagiri F, Chang HS, Eulgem T, Mauch F,

Luan S, Zou G, Whitham SA (2002) Expression profile matrix of Arabidopsis transcription factor genes suggests their putative functions in response to environmental stresses. The Plant Cell Online 14: 559

Chew O, Whelan J, Millar AH (2003) Molecular definition of the ascorbate-glutathione cycle in Arabidopsis mitochondria reveals dual targeting of antioxidant defenses in plants. Journal of Biological Chemistry 278: 46869

Chow BY, Sanchez SE, Breton G, Pruneda-Paz JL, Krogan NT, Kay SA (2014) Transcriptional regulation of LUX by CBF1 mediates cold input to the circadian clock in Arabidopsis. Current Biology 24: 1518-1524

Clifton R, Lister R, Parker KL, Sappl PG, Elhafez D, Millar AH, Day DA, Whelan J (2005) Stress-induced co-expression of alternative respiratory chain components in Arabidopsis thaliana. Plant molecular biology 58: 193-212

Clough SJ, Bent AF (1998) Floral dip: a simplified method forAgrobacterium-mediated transformation ofArabidopsis thaliana. The Plant Journal 16: 735-743

Cutler S, McCourt P (2005) Dude, where's my phenotype? Dealing with redundancy in signaling networks. Plant physiology 138: 558-559

Czechowski T, Stitt M, Altmann T, Udvardi MK, Scheible WR (2005) Genome-wide identification and testing of superior reference genes for transcript normalization in Arabidopsis. Plant physiology 139: 5

Davis BJ, Ornstein L (1959) A new high resolution electrophoresis method. Society for the Study of Blood, New York Academy of Medicine 24

Day D, Millar A, Whelan J (2004) Plant mitochondria: from genome to function. Springer Netherlands

Chapter 5 General Discussion

145

Day DA, Neuburger M, Douce R (1985) Biochemical characterization of chlorophyll-free mitochondria from pea leaves. Aust J Plant Physiol 12: 219-228

De Caluwé J, Xiao Q, Hermans C, Verbruggen N, Leloup J-C, Gonze D (2016) A Compact Model for the Complex Plant Circadian Clock. Frontiers in plant science 7

De Clercq I, Vermeirssen V, Van Aken O, Vandepoele K, Murcha MW, Law SR, Inzé A, Ng S, Ivanova A, Rombaut D (2013) The membrane-bound NAC transcription factor ANAC013 functions in mitochondrial retrograde regulation of the oxidative stress response in Arabidopsis. The Plant Cell 25: 3472-3490

de Duve C (2007) The origin of eukaryotes: a reappraisal. Nature Reviews Genetics 8: 395-403

Dimitrov L, Lam SK, Schekman R (2013) The role of the endoplasmic reticulum in peroxisome biogenesis. Cold Spring Harbor perspectives in biology 5: a013243

Dohmann EM, Kuhnle C, Schwechheimer C (2005) Loss of the CONSTITUTIVE PHOTOMORPHOGENIC9 signalosome subunit 5 is sufficient to cause the cop/det/fus mutant phenotype in Arabidopsis. The Plant Cell 17: 1967-1978

Dong S, Adams KL (2011) Differential contributions to the transcriptome of duplicated genes in response to abiotic stresses in natural and synthetic polyploids. new Phytologist 190: 1045-1057

Duncan O, Murcha MW, Whelan J (2013) Unique components of the plant mitochondrial protein import apparatus. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research 1833: 304-313

Dutilleul C, Driscoll S, Cornic G, De Paepe R, Foyer CH, Noctor G (2003) Functional mitochondrial complex I is required by tobacco leaves for optimal photosynthetic performance in photorespiratory conditions and during transients. Plant physiology 131: 264

Dyall S, Brown M, Johnson P (2004) Ancient invasions: from endosymbionts to organelles. Science 304: 253

Elkon R, Linhart C, Sharan R, Shamir R, Shiloh Y (2003) Genome-wide in silico identification of transcriptional regulators controlling the cell cycle in human cells. Genome Research 13: 773

Emelyanov VV (2001) Rickettsiaceae, rickettsia-like endosymbionts, and the origin of mitochondria. Bioscience Reports 21: 1-17

Erb M, Meldau S, Howe GA (2012) Role of phytohormones in insect-specific plant reactions. Trends in plant science 17: 250-259

Escobar MA, Franklin KA, Svensson ÅS, Salter MG, Whitelam GC, Rasmusson AG (2004) Light regulation of the Arabidopsis respiratory chain. Multiple discrete photoreceptor responses contribute to induction of type II NAD (P) H dehydrogenase genes. Plant physiology 136: 2710

Ettema TJ, Andersson SG (2009) The α-proteobacteria: the Darwin finches of the bacterial world. Biology letters: rsbl. 2008.0793

Eubel H, Jansch L, Braun HP (2003) New insights into the respiratory chain of plant mitochondria. Supercomplexes and a unique composition of complex II. Plant physiology 133: 274

Eulgem T, Somssich IE (2007) Networks of WRKY transcription factors in defense signaling. Current opinion in plant biology 10: 366-371

Falk A, Feys BJ, Frost LN, Jones JDG, Daniels MJ, Parker JE (1999) EDS1, an essential component of R gene-mediated disease resistance in Arabidopsis has homology to eukaryotic lipases. Proceedings of the National Academy of Sciences of the United States of America 96: 3292

Fernandes JC, Goulao LF, Amâncio S (2016) Regulation of cell wall remodeling in grapevine (Vitis vinifera L.) callus under individual mineral stress deficiency. Journal of plant physiology 190: 95-105

Chapter 5 General Discussion

146

Fitch WM (1970) Distinguishing homologous from analogous proteins. Systematic Biology 19: 99

Foyer CH, Noctor G (2005) Redox homeostasis and antioxidant signaling: a metabolic interface between stress perception and physiological responses. The Plant Cell Online 17: 1866

Frey TG, Mannella CA (2000) The internal structure of mitochondria. Trends in Biochemical Sciences 25: 319-324

Frey TG, Renken CW, Perkins GA (2002) Insight into mitochondrial structure and function from electron tomography. Biochimica et Biophysica Acta (BBA)-Bioenergetics 1555: 196-203

Fujisawa T, Okamoto S, Katayama T, Nakao M, Yoshimura H, Kajiya-Kanegae H, Yamamoto S, Yano C, Yanaka Y, Maita H (2014) CyanoBase and RhizoBase: databases of manually curated annotations for cyanobacterial and rhizobial genomes. Nucleic acids research 42: D666-D670

Fujita M, Fujita Y, Noutoshi Y, Takahashi F, Narusaka Y, Yamaguchi-Shinozaki K, Shinozaki K (2006) Crosstalk between abiotic and biotic stress responses: a current view from the points of convergence in the stress signaling networks. Current opinion in plant biology 9: 436-442

Fukushima A, Kusano M, Mejia RF, Iwasa M, Kobayashi M, Hayashi N, Watanabe-Takahashi A, Narisawa T, Tohge T, Hur M (2014) Metabolomic characterization of knockout mutants in Arabidopsis: development of a metabolite profiling database for knockout mutants in Arabidopsis. Plant physiology 165: 948-961

Gadjev I, Vanderauwera S, Gechev TS, Laloi C, Minkov IN, Shulaev V, Apel K, Inze D, Mittler R, Van Breusegem F (2006) Transcriptomic footprints disclose specificity of reactive oxygen species signaling in Arabidopsis. Plant physiology 141: 436

Gaspar T, Franck T, Bisbis B, Kevers C, Jouve L, Hausman JF, Dommes J (2002) Concepts in plant stress physiology. Application to plant tissue cultures. Plant Growth Regulation 37: 263-285

Gentry M, Hennig L (2014) Remodelling chromatin to shape development of plants. Experimental cell research 321: 40-46

Giraud E, Ho LHM, Clifton R, Carroll A, Estavillo G, Tan YF, Howell KA, Ivanova A, Pogson BJ, Millar AH (2008) The absence of ALTERNATIVE OXIDASE1a in Arabidopsis results in acute sensitivity to combined light and drought stress. Plant physiology 147: 595

Giraud E, Van Aken O (2012) REDOX regulation of mitochondrial function in plants. Plant, Cell & Environment

Giraud E, Van Aken O, Ho LHM, Whelan J (2009) The Transcription Factor ABI4 Is a Regulator of Mitochondrial Retrograde Expression of ALTERNATIVE OXIDASE1a1 [C][W][OA].

Giraud E, Van Aken O, Uggalla V, Whelan J (2012) REDOX regulation of mitochondrial function in plants. Plant, cell & environment 35: 271-280

Giraudat J, Hauge BM, Valon C, Smalle J, Parcy F, Goodman HM (1992) Isolation of the Arabidopsis ABI3 gene by positional cloning. The Plant Cell Online 4: 1251

Glaser E, Sjöling S, Tanudji M, Whelan J (1998) Mitochondrial protein import in plants–signals, sorting, targeting, processing and regulation. Plant molecular biology 38: 311-338

Glazebrook J, Ausubel FM (1994) Isolation of phytoalexin-deficient mutants of Arabidopsis thaliana and characterization of their interactions with bacterial pathogens. Proceedings of the National Academy of Sciences 91: 8955

Glazebrook J, Chen W, Estes B, Chang HS, Nawrath C, Métraux JP, Zhu T, Katagiri F (2003) Topology of the network integrating salicylate and jasmonate signal

Chapter 5 General Discussion

147

transduction derived from global expression phenotyping. The Plant Journal 34: 217-228

Gray JC, Sullivan JA, Wang J-H, Jerome CA, MacLean D (2003) Coordination of plastid and nuclear gene expression. Philosophical Transactions of the Royal Society of London B: Biological Sciences 358: 135-145

Gray M, Burger G, Lang B (1999) Mitochondrial evolution. Science 283: 1476 Gray MW (1989) The evolutionary origins of organelles. Trends in Genetics 5: 294-299 Gu Z, Rifkin SA, White KP, Li WH (2004) Duplicate genes increase gene expression

diversity within and between species. Nature Genetics 36: 577-579 Gu Z, Steinmetz LM, Gu X, Scharfe C, Davis RW, Li W-H (2003) Role of duplicate

genes in genetic robustness against null mutations. Nature 421: 63-66 Hamel P, Saint‐Georges Y, De Pinto B, Lachacinski N, Altamura N, Dujardin G

(2004) Redundancy in the function of mitochondrial phosphate transport in Saccharomyces cerevisiae and Arabidopsis thaliana. Molecular microbiology 51: 307-317

Han SK, Wu MF, Cui S, Wagner D (2015) Roles and activities of chromatin remodeling ATPases in plants. The Plant Journal 83: 62-77

Heath MC (2000) Hypersensitive response-related death. Plant Mol Biol 44: 321-334 Hedges S, Chen H, Kumar S, Wang D, Thompson A, Watanabe H (2001) A genomic

timescale for the origin of eukaryotes. BMC Evolutionary Biology 1: 4 Hedtke B, Wagner I, Börner T, Hess WR (1999) Inter organellar crosstalk in higher

plants: impaired chloroplast development affects mitochondrial gene and transcript levels. The Plant Journal 19: 635-643

Higo K, Ugawa Y, Iwamoto M, Korenaga T (1999) Plant cis-acting regulatory DNA elements (PLACE) database: 1999. Nucleic acids research 27: 297

Ho LHM, Giraud E, Uggalla V, Lister R, Clifton R, Glen A, Thirkettle-Watts D, Van Aken O, Whelan J (2008) Identification of regulatory pathways controlling gene expression of stress-responsive mitochondrial proteins in Arabidopsis. Plant physiology 147: 1858

Holtorf H, Guitton MC, Reski R (2002) Plant functional genomics. Naturwissenschaften 89: 235-249

Hooper CM, Tanz SK, Castleden IR, Vacher MA, Small ID, Millar AH (2014) SUBAcon: a consensus algorithm for unifying the subcellular localization data of the Arabidopsis proteome. Bioinformatics 30: 3356-3364

Howell KA, Millar AH, Whelan J (2006) Ordered assembly of mitochondria during rice germination begins with promitochondrial structures rich in components of the protein import apparatus. Plant molecular biology 60: 201-223

Howell KA, Millar AH, Whelan J (2007) Building the Powerhouse: What are the Signals Involved in Plant Mitochondrial Biogenesis? Plant Signaling & Behavior 2: 428

Hruz T, Laule O, Szabo G, Wessendorp F, Bleuler S, Oertle L, Widmayer P, Gruissem W, Zimmermann P (2008) Genevestigator v3: a reference expression database for the meta-analysis of transcriptomes. Advances in bioinformatics 2008

Huang J, Cardoza Y, Schmelz E, Raina R, Engelberth J, Tumlinson J (2003) Differential volatile emissions and salicylic acid levels from tobacco plants in response to different strains of <i>Pseudomonas syringae</i>. Planta 217: 767

Huang S, Van Aken O, Schwarzländer M, Belt K, Millar AH (2016) The roles of mitochondrial reactive oxygen species in cellular signaling and stress response in plants. Plant physiology 171: 1551-1559

Iwamoto K, Bundo M, Kato T (2005) Altered expression of mitochondria-related genes in postmortem brains of patients with bipolar disorder or schizophrenia, as revealed by large-scale DNA microarray analysis. Human molecular genetics 14: 241

Chapter 5 General Discussion

148

Jaeger KE, Wigge PA (2007) FT protein acts as a long-range signal in Arabidopsis. Current Biology 17: 1050-1054

Jefferson RA, Kavanagh TA, Bevan MW (1987) GUS fusions: beta-glucuronidase as a sensitive and versatile gene fusion marker in higher plants. The EMBO Journal 6: 3901

Jiao S, Thornsberry JM, Elthon TE, Newton KJ (2005) Biochemical and molecular characterization of photosystem I deficiency in the NCS6 mitochondrial mutant of maize. Plant molecular biology 57: 303-313

Jin Z, Xue S, Luo Y, Tian B, Fang H, Li H, Pei Y (2013) Hydrogen sulfide interacting with abscisic acid in stomatal regulation responses to drought stress in Arabidopsis. Plant Physiology and Biochemistry 62: 41-46

Karimi M, Inzé D, Depicker A (2002) GATEWAY™ vectors for Agrobacterium-mediated plant transformation. Trends in plant science 7: 193-195

Kim E, Ok J, Lee S, Kim J, Kwon E, Lim D, Choi S, Lee S (2014) Effects of Anthelmintic Plant Extracts on Ruminal Fermentation Characteristics, Bacterial Diversity and Methane Production in vitro. Journal of Agriculture & Life Science 48: 113-120

Kim HS, Yu Y, Snesrud EC, Moy LP, Linford LD, Haas BJ, Nierman WC, Quackenbush J (2005) Transcriptional divergence of the duplicated oxidative stress-responsive genes in the Arabidopsis genome. The Plant Journal 41: 212-220

Kleffmann T, Russenberger D, von Zychlinski A, Christopher W, Sjölander K, Gruissem W, Baginsky S (2004) The Arabidopsis thaliana chloroplast proteome reveals pathway abundance and novel protein functions. Current Biology 14: 354-362

Kleine T, Leister D (2016) Retrograde signaling: organelles go networking. Biochimica Et Biophysica Acta (BBA)-Bioenergetics 1857: 1313-1325

Koo AJK, Gao X, Jones AD, Howe GA (2009) A rapid wound signal activates the systemic synthesis of bioactive jasmonates in Arabidopsis. Plant J 59: 974-986

Koornneef A, Pieterse CMJ (2008) Cross talk in defense signaling. Plant physiology 146: 839

Kotera E, Tasaka M, Shikanai T (2005) A pentatricopeptide repeat protein is essential for RNA editing in chloroplasts. Nature 433: 326-330

Koussevitzky S, Nott A, Mockler TC, Hong F, Sachetto-Martins G, Surpin M, Lim J, Mittler R, Chory J (2007) Signals from chloroplasts converge to regulate nuclear gene expression. Science 316: 715

Krause M, Durner J (2004) Harpin inactivates mitochondria in Arabidopsis suspension cells. Molecular plant-microbe interactions 17: 131-139

Küchler M, Decker S, Hörmann F, Soll J, Heins L (2002) Protein import into chloroplasts involves redox-regulated proteins. The EMBO Journal 21: 6136-6145

Kusano T, Tateda C, Berberich T, Takahashi Y (2009) Voltage-dependent anion channels: their roles in plant defense and cell death. Plant cell reports 28: 1301-1308

Kwong JQ, Beal MF, Manfredi G (2006) The role of mitochondria in inherited neurodegenerative diseases. Journal of neurochemistry 97: 1659-1675

Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA, McWilliam H, Valentin F, Wallace IM, Wilm A, Lopez R (2007) Clustal W and Clustal X version 2.0. Bioinformatics 23: 2947

Law SR, Narsai R, Whelan J (2014) Mitochondrial biogenesis in plants during seed germination. Mitochondrion 19: 214-221

Lawton K, Weymann K, Friedrich L, Vemooij B, Uknes S, Ryals J (1995) Systemic acquired resistance in Arabidopsis requires salicylic acid but not ethylene. Molecular Plant Microbe Interactions 8: 863-870

Chapter 5 General Discussion

149

Lee MN, Whelan J (2004) Identification of signals required for import of the soybean F(A)d subunit of ATP synthase into mitochondria. Plant Mol Biol 54: 193-203

Leister D (2005) Genomics-based dissection of the cross-talk of chloroplasts with the nucleus and mitochondria in Arabidopsis. Gene 354: 110-116

Li N, Ragheb K, Lawler G, Sturgis J, Rajwa B, Melendez JA, Robinson JP (2003) Mitochondrial complex I inhibitor rotenone induces apoptosis through enhancing mitochondrial reactive oxygen species production. Journal of Biological Chemistry 278: 8516

Lindemann P, Koch A, Degenhardt B, Hause G, Grimm B, Papadopoulos V (2004) A novel Arabidopsis thaliana protein is a functional peripheral-type benzodiazepine receptor. Plant and cell physiology 45: 723

Lister R, Carrie C, Duncan O, Ho LH, Howell KA, Murcha MW, Whelan J (2007) Functional definition of outer membrane proteins involved in preprotein import into mitochondria. The Plant Cell 19: 3739-3759

Lister R, Carrie C, Duncan O, Ho LH, Howell KA, Murcha MW, Whelan J (2007) Functional definition of outer membrane proteins involved in preprotein import into mitochondria. Plant Cell 19: 3739-3759

Lister R, Chew O, Lee MN, Heazlewood JL, Clifton R, Parker KL, Millar AH, Whelan J (2004) A transcriptomic and proteomic characterization of the Arabidopsis mitochondrial protein import apparatus and its response to mitochondrial dysfunction. Plant physiology 134: 777

Lister R, Chew O, Rudhe C, Lee M-N, Whelan J (2001) Arabidopsis thaliana

ferrochelatase‐I and‐II are not imported into Arabidopsis mitochondria. FEBS letters 506: 291-295

Lister R, Gregory BD, Ecker JR (2009) Next is now: new technologies for sequencing of genomes, transcriptomes, and beyond. Current opinion in plant biology 12: 107-118

Liu L, White MJ, MacRae TH (1999) Transcription factors and their genes in higher plants. European Journal of Biochemistry 262: 247-257

Liu Z, Butow RA (2006) Mitochondrial retrograde signaling. Annu. Rev. Genet. 40: 159-185

Lynch M, Conery JS (2000) The evolutionary fate and consequences of duplicate genes. Science 290: 1151

Mackenzie S, McIntosh L (1999) Higher plant mitochondria. The Plant Cell Online 11: 571

Martin DB, Vagelos PR (1962) The mechanism of tricarboxylic acid cycle regulation of fatty acid synthesis. Journal of Biological Chemistry 237: 1787

Martin W, Müller M (1998) The hydrogen hypothesis for the first eukaryote. Nature 392: 37-41

Maxwell DP, Nickels R, McIntosh L (2002) Evidence of mitochondrial involvement in the transduction of signals required for the induction of genes associated with pathogen attack and senescence. The Plant Journal 29: 269-279

Maxwell DP, Wang Y, McIntosh L (1999) The alternative oxidase lowers mitochondrial reactive oxygen production in plant cells. Proceedings of the National Academy of Sciences of the United States of America 96: 8271

McFadden GI (1999) Plastids and Protein Targeting1. Journal of Eukaryotic Microbiology 46: 339-346

Meskauskiene R, Nater M, Goslings D, Kessler F, op den Camp R, Apel K (2001) FLU: a negative regulator of chlorophyll biosynthesis in Arabidopsis thaliana. Proceedings of the national academy of sciences 98: 12826-12831

Meyuhas O, Perry RP (1979) Relationship between size, stability and abundance of the messenger RNA of mouse L cells. Cell 16: 139-148

Mi H, Huang X, Muruganujan A, Tang H, Mills C, Kang D, Thomas PD (2016) PANTHER version 11: expanded annotation data from Gene Ontology and

Chapter 5 General Discussion

150

Reactome pathways, and data analysis tool enhancements. Nucleic Acids Research: gkw1138

Millar A (2007) The plant mitochondrial proteome. Plant Proteomics: 226-246 Millar AH, Leaver CJ (2000) The cytotoxic lipid peroxidation product, 4-hydroxy-2-

nonenal, specifically inhibits decarboxylating dehydrogenases in the matrix of plant mitochondria. FEBS letters 481: 117-121

Millar AH, Sweetlove LJ, Giegé P, Leaver CJ (2001) Analysis of the Arabidopsis mitochondrial proteome. Plant physiology 127: 1711

Millar AH, Whelan J, Soole KL, Day DA (2011) Organization and regulation of mitochondrial respiration in plants. Annual review of plant biology 62: 79-104

Millar H, Considine MJ, Day DA, Whelan J (2001) Unraveling the role of mitochondria during oxidative stress in plants. IUBMB life 51: 201-205

Misra RC, Sandeep MK, Kumar S, Ghosh S (2016) A thaumatin-like protein of Ocimum basilicum confers tolerance to fungal pathogen and abiotic stress in transgenic Arabidopsis. Scientific reports 6

Mitchell P, Moyle J (1967) Chemiosmotic hypothesis of oxidative phosphorylation. Nature 213: 137-139

Moore AL, Albury MS, Crichton PG, Affourtit C (2002) Function of the alternative oxidase: is it still a scavenger? Trends in plant science 7: 478-481

Murcha MW, Elhafez D, Lister R, Tonti-Filippini J, Baumgartner M, Philippar K, Carrie C, Mokranjac D, Soll J, Whelan J (2007) Characterization of the preprotein and amino acid transporter gene family in Arabidopsis. Plant physiology 143: 199

Murcha MW, Elhafez D, Lister R, Tonti-Filippini J, Baumgartner M, Philippar K, Carrie C, Mokranjac D, Soll J, Whelan J (2007) Characterization of the preprotein and amino acid transporter gene family in Arabidopsis. Plant Physiol 143: 199-212

Murcha MW, Huang T, Whelan J (1999) Import of precursor proteins into mitochondria from soybean tissues during development. FEBS letters 464: 53-59

Murcha MW, Wang Y, Narsai R, Whelan J (2014) The plant mitochondrial protein import apparatus—the differences make it interesting. Biochimica et Biophysica Acta (BBA)-General Subjects 1840: 1233-1245

Mustroph A, KAISER KA, LARIVE CK, BAILEY‐SERRES J (2014) Characterization

of distinct root and shoot responses to low‐oxygen stress in Arabidopsis with a focus on primary C‐and N‐metabolism. Plant, cell & environment 37: 2366-2380

Nakashima K, Yamaguchi-Shinozaki K, Shinozaki K (2014) The transcriptional regulatory network in the drought response and its crosstalk in abiotic stress responses including drought, cold, and heat. Abiotic Stress: Molecular Genetics and Genomics: 25

Navrot N, Collin V, Gualberto J, Gelhaye E, Hirasawa M, Rey P, Knaff DB, Issakidis E, Jacquot JP, Rouhier N (2006) Plant glutathione peroxidases are functional peroxiredoxins distributed in several subcellular compartments and regulated during biotic and abiotic stresses. Plant physiology 142: 1364

Ng S, De Clercq I, Van Aken O, Law SR, Ivanova A, Willems P, Giraud E, Van Breusegem F, Whelan J (2014) Anterograde and retrograde regulation of nuclear genes encoding mitochondrial proteins during growth, development, and stress. Molecular plant 7: 1075-1093

Nisoli E, Clementi E, Moncada S, Carruba M (2004) Mitochondrial biogenesis as a cellular signaling framework. Biochemical pharmacology 67: 1-15

Norman C, Howell KA, Millar AH, Whelan JM, Day DA (2004) Salicylic acid is an uncoupler and inhibitor of mitochondrial electron transport. Plant Physiology 134: 492-501

Chapter 5 General Discussion

151

O'Connor TR, Dyreson C, Wyrick JJ (2005) Athena: a resource for rapid visualization and systematic analysis of Arabidopsis promoter sequences. Bioinformatics 21: 4411-4413

Ordog SH, Higgins VJ, Vanlerberghe GC (2002) Mitochondrial alternative oxidase is not a critical component of plant viral resistance but may play a role in the hypersensitive response. Plant physiology 129: 1858

Osteryoung KW, Nunnari J (2003) The division of endosymbiotic organelles. Science 302: 1698

Palaniswamy SK, James S, Sun H, Lamb RS, Davuluri RV, Grotewold E (2006) AGRIS and AtRegNet. a platform to link cis-regulatory elements and transcription factors into regulatory networks. Plant physiology 140: 818-829

Palenik B (2002) The genomics of symbiosis: Hosts keep the baby and the bath water. Proceedings of the National Academy of Sciences of the United States of America 99: 11996

Palmieri L, Picault N, Arrigoni R, Besin E, Palmieri F, Hodges M (2008) Molecular identification of three Arabidopsis thaliana mitochondrial dicarboxylate carrier isoforms: organ distribution, bacterial expression, reconstitution into liposomes and functional characterization. Biochemical Journal 410: 621-629

Paranjape K, Hallenbeck PC (2015) Microalgal Production of Hydrogen and Biodiesel. Marine Bioenergy: Trends and Developments: 371

Parihar MS, Brewer GJ (2007) Simultaneous age related depolarization of mitochondrial membrane potential and increased mitochondrial reactive oxygen species production correlate with age related glutamate excitotoxicity in rat hippocampal neurons. Journal of neuroscience research 85: 1018-1032

Paschen SA, Rothbauer U, Káldi K, Bauer MF, Neupert W, Brunner M (2000) The role of the TIM8–13 complex in the import of Tim23 into mitochondria. The EMBO journal 19: 6392-6400

Pédelacq JD, Cabantous S, Tran T, Terwilliger TC, Waldo GS (2005) Engineering and characterization of a superfolder green fluorescent protein. Nature biotechnology 24: 79-88

Phillips GJ (2001) Green fluorescent protein–a bright idea for the study of bacterial protein localization. FEMS microbiology letters 204: 9-18

Plattner H, Salpeter M, Saltzgaber J, Schatz G (1970) Promitochondria of anaerobically grown yeast, IV. Conversion into respiring mitochondria. Proceedings of the National Academy of Sciences of the United States of America 66: 1252

Pollard TD (2003) Biochemistry A Clinical Companion to Accompany Biochemistry. JAMA: The Journal of the American Medical Association 290: 1648

Poole AM, Penny D (2007) Evaluating hypotheses for the origin of eukaryotes. Bioessays 29: 74-84

Quirós PM, Mottis A, Auwerx J (2016) Mitonuclear communication in homeostasis and stress. Nature reviews Molecular cell biology 17: 213-226

Raghavendra AS, Padmasree K (2003) Beneficial interactions of mitochondrial metabolism with photosynthetic carbon assimilation. Trends in Plant Science 8: 546-553

Rasmusson AG, Soole KL, Elthon TE (2004) Alternative NAD (P) H dehydrogenases of plant mitochondria. Plant Biology 55: 23

Rhoads DM, Subbaiah CC (2007) Mitochondrial retrograde regulation in plants. Mitochondrion 7: 177-194

Rhoads DM, Umbach AL, Subbaiah CC, Siedow JN (2006) Mitochondrial reactive oxygen species. Contribution to oxidative stress and interorganellar signaling. Plant physiology 141: 357

Robles P, Micol J (2001) Genome-wide linkage analysis of Arabidopsis genes required for leaf development. Molecular Genetics and Genomics 266: 12-19

Chapter 5 General Discussion

152

Robson CA, Vanlerberghe GC (2002) Transgenic plant cells lacking mitochondrial alternative oxidase have increased susceptibility to mitochondria-dependent and-independent pathways of programmed cell death. Science's STKE 129: 1908

Roessner U, Luedemann A, Brust D, Fiehn O, Linke T, Willmitzer L, Fernie AR (2001) Metabolic profiling allows comprehensive phenotyping of genetically or environmentally modified plant systems. The Plant Cell Online 13: 11

Rombauts S, Déhais P, Van Montagu M, Rouzé P (1999) PlantCARE, a plant cis-acting regulatory element database. Nucleic acids research 27: 295-296

Rosenfeld E, Schaeffer J, Beauvoit B, Salmon J (2004) Isolation and properties of promitochondria from anaerobic stationary-phase yeast cells. Antonie Van Leeuwenhoek 85: 9-21

Rosner BA (2006) Fundamentals of biostatistics. Duxbury Resource Center Ruan Y, Gilmore J, Conner T (1998) Towards Arabidopsis genome analysis:

monitoring expression profiles of 1400 genes using cDNA microarrays. The Plant Journal 15: 821-833

Rushton PJ, Somssich IE, Ringler P, Shen QJ (2010) WRKY transcription factors. Trends in plant science 15: 247-258

Sabar M, De Paepe R, De Kouchkovsky Y (2000) Complex I impairment, respiratory compensations, and photosynthetic decrease in nuclear and mitochondrial male sterile mutants of Nicotiana sylvestris. Plant physiology 124: 1239

Saitou N, Nei M (1987) The neighbor-joining method: a new method for reconstructing phylogenetic trees. Molecular biology and evolution 4: 406

Sanger F, Nicklen S, Coulson AR (1977) DNA sequencing with chain-terminating inhibitors. Proceedings of the National Academy of Sciences of the United States of America 74: 5463

Sänger N, Strohmeier R, Kaufmann M, Kuhl H (2000) Cell cycle-related expression and ligand binding of peripheral benzodiazepine receptor in human breast cancer cell lines. European Journal of Cancer 36: 2157-2163

Sato S, Nakamura Y, Kaneko T, Asamizu E, Tabata S (1999) Complete structure of the chloroplast genome of Arabidopsis thaliana. DNA Research 6: 283

Schägger H, von Jagow G (1991) Blue native electrophoresis for isolation of membrane protein complexes in enzymatically active form. Analytical biochemistry 199: 223-231

Schleiff E, Becker T (2010) Common ground for protein translocation: access control for mitochondria and chloroplasts. Nature Reviews Molecular Cell Biology 12: 48-59

Schwab R, Ossowski S, Riester M, Warthmann N, Weigel D (2006) Highly specific gene silencing by artificial microRNAs in Arabidopsis. The Plant Cell Online 18: 1121

Schwarzländer M, König A-C, Sweetlove LJ, Finkemeier I (2011) The impact of impaired mitochondrial function on retrograde signalling: a meta-analysis of transcriptomic responses. Journal of experimental botany 63: 1735-1750

Senkler J, Senkler M, Eubel H, Hildebrandt T, Lengwenus C, Schertl P, Schwarzländer M, Wagner S, Wittig I, Braun HP (2016) The mitochondrial complexome of Arabidopsis thaliana. The Plant Journal

Shah J (2003) The salicylic acid loop in plant defense. Current opinion in plant biology 6: 365-371

Shaughnessy DT, McAllister K, Worth L, Haugen AC, Meyer JN, Domann FE, Van Houten B, Mostoslavsky R, Bultman SJ, Baccarelli AA (2014) Mitochondria, energetics, epigenetics, and cellular responses to stress. Environmental Health Perspectives (Online) 122: 1271

Chapter 5 General Discussion

153

Shinozaki K, Yamaguchi-Shinozaki K, Seki M (2003) Regulatory network of gene expression in the drought and cold stress responses. Curr Opin Plant Biol 6: 410-417

Siedow J, Umbach A (1995) Plant mitochondrial electron transfer and molecular biology. The Plant Cell Online 7: 821

Singh K, Foley RC, Onate-Sanchez L (2002) Transcription factors in plant defense and stress responses. Curr Opin Plant Biol 5: 430-436

Sirrenberg C, Bauer MF, Guiard B, Neupert W, Brunner M (1996) Import of carrier proteins into the mitochondrial inner membrane mediated by Tim22.

Skriver K, Mundy J (1990) Gene expression in response to abscisic acid and osmotic stress. Plant Cell 2: 503-512

Slonim DK, Yanai I (2009) Getting started in gene expression microarray analysis. PLoS computational biology 5

Sohn KH, Lee SC, Jung HW, Hong JK, Hwang BK (2006) Expression and functional roles of the pepper pathogen-induced transcription factor RAV1 in bacterial disease resistance, and drought and salt stress tolerance. Plant molecular biology 61: 897-915

Soll J, Schleiff E (2004) Protein import into chloroplasts. Nature Reviews Molecular Cell Biology 5: 198-208

Staswick PE, Su W, Howell SH (1992) Methyl jasmonate inhibition of root growth and induction of a leaf protein are decreased in an Arabidopsis thaliana mutant. Proceedings of the National Academy of Sciences of the United States of America 89: 6837

Stefano GB, Kream RM (2016) Glycolytic Coupling to Mitochondrial Energy Production Ensures Survival in an Oxygen Rich Environment. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research 22: 2571

Steffens NO, Galuschka C, Schindler M, Bülow L, Hehl R (2004) AthaMap: an online resource for in silico transcription factor binding sites in the Arabidopsis thaliana genome. Nucleic acids research 32: D368-D372

Sticher L, Mauch-Mani B, Metraux JP (1997) Systemic acquired resistance. Annu Rev Phytopathology 35: 235-270

Sun Y, Zhang J, Lu L, Chen SS, Quinn MT, Weber KT (2002) Aldosterone-induced inflammation in the rat heart: role of oxidative stress. American Journal of Pathology 161: 1773

Suter L, Widmer A (2013) Phenotypic effects of salt and heat stress over three generations in Arabidopsis thaliana.

Suzuki N, Koussevitzky S, Mittler R, Miller G (2012) ROS and redox signalling in the response of plants to abiotic stress. Plant, Cell & Environment 35: 259-270

Swamy M, Siegers GM, Minguet S, Wollscheid B, Schamel WW (2006) Blue native polyacrylamide gel electrophoresis (BN-PAGE) for the identification and analysis of multiprotein complexes. Sci. STKE 2006: pl4-pl4

Swarbreck D, Wilks C, Lamesch P, Berardini TZ, Garcia-Hernandez M, Foerster H, Li D, Meyer T, Muller R, Ploetz L (2007) The Arabidopsis Information Resource (TAIR): gene structure and function annotation. Nucleic Acids Research

Sweetlove LJ, Heazlewood JL, Herald V, Holtzapffel R, Day DA, Leaver CJ, Millar AH (2002) The impact of oxidative stress on Arabidopsis mitochondria. The Plant Journal 32: 891-904

Tanz SK, Castleden I, Hooper CM, Vacher M, Small I, Millar HA (2012) SUBA3: a database for integrating experimentation and prediction to define the SUBcellular location of proteins in Arabidopsis. Nucleic acids research: gks1151

Taylor NL, Day DA, Millar AH (2004) Targets of stress induced oxidative damage in plant mitochondria and their impact on cell carbon/nitrogen metabolism. Journal of experimental botany 55: 1

Chapter 5 General Discussion

154

Taylor NL, Heazlewood JL, Day DA, Millar AH (2004) Lipoic acid-dependent oxidative catabolism of {alpha}-keto acids in mitochondria provides evidence for branched-chain amino acid catabolism in Arabidopsis. Plant physiology 134: 838

Taylor NL, Rudhe C, Hulett JM, Lithgow T, Glaser E, Day DA, Millar AH, Whelan J (2003) Environmental stresses inhibit and stimulate different protein import pathways in plant mitochondria. FEBS letters 547: 125-130

Teige M, Scheikl E, Eulgem T, Doczi R, Ichimura K, Shinozaki K, Dangl JL, Hirt H (2004) The MKK2 pathway mediates cold and salt stress signaling in Arabidopsis. Mol Cell 15: 141-152

Terziyska N, Lutz T, Kozany C, Mokranjac D, Mesecke N, Neupert W, Herrmann JM, Hell K (2005) Mia40, a novel factor for protein import into the intermembrane space of mitochondria is able to bind metal ions. FEBS letters 579: 179-184

Thirkettle-Watts D, McCabe TC, Clifton R, Moore C, Finnegan PM, Day DA, Whelan J (2003) Analysis of the alternative oxidase promoters from soybean. Plant Physiol 133: 1158-1169

Uggalla V (2006) Expression analysis and functional characterisation of the stress induced gene UPOX1, a gene encoded by At2g21640 in Arabidopsis. Honours. University of Western Australia, Perth

Unseld M, Marienfeld JR, Brandt P, Brennicke A (1997) The mitochondrial genome of Arabidopsis thaliana contains 57 genes in 366,924 nucleotides. Nature Genetics 15: 57-61

Vacca RA, de Pinto MC, Valenti D, Passarella S, Marra E, De Gara L (2004) Production of reactive oxygen species, alteration of cytosolic ascorbate peroxidase, and impairment of mitochondrial metabolism are early events in heat shock-induced programmed cell death in tobacco Bright-Yellow 2 cells. Plant physiology 134: 1100

Van Aken O, De Clercq I, Law SR, Van Breusegem F, Millar AH, Whelan J (2016) Mitochondrial and chloroplast stress responses are modulated in distinct touch and chemical inhibition phases in Arabidopsis. Plant physiology: pp. 00273.02016

Van Aken O, Ford E, Lister R, Huang S, Millar AH (2016) Retrograde signalling caused by heritable mitochondrial dysfunction is partially mediated by ANAC017 and improves plant performance. The Plant Journal

Van Aken O, Giraud E, Clifton R, Whelan J (2009) Alternative oxidase: a target and regulator of stress responses. Physiologia plantarum 137: 354-361

Van Aken O, Pečenková T, Van De Cotte B, De Rycke R, Eeckhout D, Fromm H, De Jaeger G, Witters E, Beemster GT, Inzé D (2007) Mitochondrial type‐I prohibitins of Arabidopsis thaliana are required for supporting proficient meristem development. The Plant Journal 52: 850-864

Van Aken O, Van Breusegem F (2015) Licensed to kill: mitochondria, chloroplasts, and cell death. Trends in plant science 20: 754-766

Van Aken O, Whelan J (2012) Comparison of transcriptional changes to chloroplast and mitochondrial perturbations reveals common and specific responses in Arabidopsis. Frontiers in plant science 3

Van Aken O, Whelan J, Van Breusegem F (2010) Prohibitins: mitochondrial partners in development and stress response. Trends in plant science 15: 275-282

Van Aken O, Zhang B, Carrie C, Uggalla V, Paynter E, Giraud E, Whelan J (2009) Defining the Mitochondrial Stress Response in Arabidopsis thaliana. Molecular plant

Van Aken O, Zhang B, Law S, Narsai R, Whelan J (2013) AtWRKY40 and AtWRKY63 modulate the expression of stress-responsive nuclear genes encoding mitochondrial and chloroplast proteins. Plant physiology 162: 254-271

Chapter 5 General Discussion

155

van der Giezen M, Tovar J, Clark CG (2005) Mitochondrion-derived organelles in protists and fungi. International review of cytology 244: 175-225

van Lis R, Atteia A (2004) Control of mitochondrial function via photosynthetic redox signals. Photosynthesis Research 79: 133-148

Vanlerberghe GC (2013) Alternative oxidase: a mitochondrial respiratory pathway to maintain metabolic and signaling homeostasis during abiotic and biotic stress in plants. International journal of molecular sciences 14: 6805-6847

Vanlerberghe GC, McIntosh L (1997) Alternative oxidase: from gene to function. Annual Review of Plant Biology 48: 703-734

Vanlerberghe GC, Robson CA, Yip JYH (2002) Induction of mitochondrial alternative oxidase in response to a cell signal pathway down-regulating the cytochrome pathway prevents programmed cell death. Plant physiology 129: 1829

Verniquet F, Gaillard J, Neuburger M, Douce R (1991) Rapid inactivation of plant aconitase by hydrogen peroxide. Biochemical journal 276: 643

Villanueva L, Schouten S, Sinninghe Damsté JS (2016) Phylogenomic analysis of lipid biosynthetic genes of Archaea shed light on the ‘lipid divide’. Environmental microbiology

Vlot AC, Dempsey DMA, Klessig DF (2009) Salicylic acid, a multifaceted hormone to combat disease. Annual review of phytopathology 47: 177-206

Wang C, Krishnakumar S, Wilhelmy J, Babrzadeh F, Stepanyan L, Su LF, Levinson D, Fernandez-Viña MA, Davis RW, Davis MM (2012) High-throughput, high-fidelity HLA genotyping with deep sequencing. Proceedings of the National Academy of Sciences 109: 8676-8681

Weltmeier F, Rahmani F, Ehlert A, Dietrich K, Schütze K, Wang X, Chaban C, Hanson J, Teige M, Harter K (2009) Expression patterns within the Arabidopsis C/S1 bZIP transcription factor network: availability of heterodimerization partners controls gene expression during stress response and development. Plant molecular biology 69: 107-119

Werhahn W, Niemeyer A, Jansch L, Kruft V, Schmitz UK, Braun HP (2001) Purification and characterization of the preprotein translocase of the outer mitochondrial membrane from Arabidopsis. Identification of multiple forms of TOM20. Plant physiology 125: 943

Whelan J, Tanudji MR, Kathleen Smith M, Day DA (1996) Evidence for a link between translocation and processing during protein import into soybean mitochondria. BBA-Molecular Cell Research 1312: 48-54

White R (1979) Acetylsalicylic acid (asprin) induces resistance to tobacco mosaic virus in tobacco. Virology 99: 410-412

Wilhelm BT, Marguerat S, Watt S, Schubert F, Wood V, Goodhead I, Penkett CJ, Rogers J, Bähler J (2008) Dynamic repertoire of a eukaryotic transcriptome surveyed at single-nucleotide resolution. Nature 453: 1239-1243

Wong JH, Cai N, Balmer Y, Tanaka CK, Vensel WH, Hurkman WJ, Buchanan BB (2004) Thioredoxin targets of developing wheat seeds identified by complementary proteomic approaches. Phytochemistry 65: 1629-1640

Woodson JD, Chory J (2008) Coordination of gene expression between organellar and nuclear genomes. Nature Reviews Genetics 9: 383-395

Wray GA, Hahn MW, Abouheif E, Balhoff JP, Pizer M, Rockman MV, Romano LA (2003) The evolution of transcriptional regulation in eukaryotes. Molecular biology and evolution 20: 1377

Yanagisawa S, Sheen J (1998) Involvement of maize Dof zinc finger proteins in tissue-specific and light-regulated gene expression. The Plant Cell Online 10: 75

Yang Y, Glynn JM, Olson BJSC, Schmitz AJ, Osteryoung KW (2008) Plastid division: across time and space. Current Opinion in Plant Biology 11: 577-584

Yanhui C, Xiaoyuan Y, Kun H, Meihua L, Jigang L, Zhaofeng G, Zhiqiang L, Yunfei Z, Xiaoxiao W, Xiaoming Q (2006) The MYB transcription factor superfamily of

Chapter 5 General Discussion

156

Arabidopsis: expression analysis and phylogenetic comparison with the rice MYB family. Plant molecular biology 60: 107-124

Yoshida K, Noguchi K (2011) Interaction Between Chloroplasts and Mitochondria: Activity, Function, and Regulation of the Mitochondrial Respiratory System during Photosynthesis. Plant Mitochondria: 383-409

Yu J, Nickels R, McIntosh L (2001) A genome approach to mitochondrial-nuclear communicationin Arabidopsis. Plant physiology and biochemistry 39: 345-353

Zhang B, Van Aken O, Thatcher L, De Clercq I, Duncan O, Law SR, Murcha MW, Merwe M, Seifi HS, Carrie C (2014) The mitochondrial outer membrane AAA ATPase AtOM66 affects cell death and pathogen resistance in Arabidopsis thaliana. The Plant Journal 80: 709-727

Zhang Y, Marcillat O, Giulivi C, Ernster L, Davies KJ (1990) The oxidative inactivation of mitochondrial electron transport chain components and ATPase. Journal of Biological Chemistry 265: 16330

Appendix 1

157

Appendix 1

Plant Growth Media

Cell Culture Media

1 sachet/L Murashige & Skoog Salt Mixture (N1145; Invitrogen, Australia), 30 g/L

sucrose, 500 µl 1 mg/ml Naphthalene acetic acid (K salt), 50 µl 1 mg/ml kinetin.

Liquid Culture Media

1 sachet/L Murashige & Skoog Salt Mixture (N1145; Invitrogen, Australia), 400 µl

Gamborg’s B5 vitamins (Sigma Aldrich, Australia), 0.32 g MES, 16 g sucrose. Adjust

pH to 5.7 using KOH, and autoclave to sterilise. Add 50 µg/ml cefotaxime sodium

antibiotic (Claforan®).

Solid Culture Media

1 sachet/L Gamborg’s B5 Basal Salt mixture (G5768; Sigma Aldrich, Australia), 1 ml/L

1000X Gamborg’s B5 vitamins, 0.5 g/l MES, 30 g/L sucrose. Adjust pH to 5.7 using

KOH, add 6 g/L agar and autoclave to sterilise. Prior to pouring media, add 50 µg/ml

cefotaxime sodium antibiotic (Claforan®).

Seed Sterilization Solution

After washing the seeds in 70% ethanol a solution of 5% bleech with 0.1% Tween 20

was applied to the seeds. This solution was washed with SDW prior to sowing.

Bacterial Growth Media

SOC Media

20 g/L tryptone peptone, 5 g/L yeast extract, 10 ml/L 1 M NaCl, 2.5 ml/L KCl.

Autoclave to sterilize. Add 5 ml/L 1 M MgCl2, 5 ml/L 1 M MgSO4, 10 ml/L filter

sterilized 2 M glucose prior to use.

Luria-Bertani (LB) media

10 g/L tryptone peptone, 5 g/L yeast extract, 10 g/L NaCl, adjust pH to 7.0 using 1 M

NaOH. Sterilize by autoclaving liquid. Add 15 g/L bacto agar prior to autoclaving to

Appendix 1

158

make solid media LB plates. Add appropriate antibiotics following cooling of liquids

such as Amicillin to a final concentration of 50 g/ml prior to pouring plates.

NZY+ media

10 g/L NZ amine (casein hydrolysate), 5 g/L yeast extract, 5 g/L NaCL. Sterilize by

autoclaving. Add the following prior to use; 12.5 ml/L 1 M MgCl2, 12.5 ml/L MgSo4 and

10 ml/L 1 M filter sterilized 2 M glucose.

Agarose gel electrophoresis solutions

TAE Buffer

48 g/L Tris, 11.8 ml/L glacial acetic acid, 10 mM EDTA. Dilute 1 in 10 in ddH2O for

use as 1 x TAE.

Loading Buffer

30% (v/v) glycerol, 0.05% (v/v) bromophenol blue.

5 x Running Buffer

15 g/L Tris base, 72 g/L glycine, 5 g/L SDS. Do not adjust pH but check it it lies

between 8.3-8.5. Dilute 1 in 5 with ddH2O for use as 1 x Running Buffer.

Tris-Tricine 1 x Cathode Buffer

12.11 g/L Tris base, 17.92 g/L tricine. 1 g/L SDS. Do not adjust pH

Tris-Tricine 5 x Anode Buffer

121.1 g/L Tris base. Adjust pH to 8.9 with concentrated HCL.

Coomassie stain solution

100 ml/L glacial acetic acid, 400 ml/L 100% ethanol, 1 g/L Coomassie Brilliant Blue R-

250. Vacuum filtered to remove undissolved grains.

Appendix 1

159

Coomassie destain solution

150 ml/L glacial acetic acid, 375 ml/L 100% ethanol, 25 ml/L glycerol.

SDS-PAGE solutions

Separating buffer

1.5 M Tris-HCL, pH8.8

Stacking buffer

0.5 M Tris-HCL, pH 6.8

Separating gel (12%)

Add 5.25 ml separating buffer, 8.4 ml 30% (w/v) acrylamide/bisacrylamide and 210 µl

10% (w/v) SDS to 7 ml ddH2O. Degas via vacuum manifold for 5 min and add 120 µl

10% (w/v) ammonium persulfate and 10.5 µl TEMED just prior to use.

Separating gel (14%)

Add 5.25 ml separating buffer, 9.8 ml 30% (w/v) acrylamide/bisacrylamide and 210 µl

10% (w/v) SDS to 5.6 ml ddH2O. Degas via vacuum manifold for 5 min and add 120 µl

10% (w/v) ammonium persulfate and 10.5 µl TEMED just prior to use.

Stacking gel (4%)

Add 2.5 ml stacking buffer, 1.3 ml 30% (w/v) acrylamide/bisacrylamide and 100 µl

10% (w/v) SDS to 6.1 ml ddH2O. Degas via vacuum manifold for 5 min and add 50 µl

10% (w/v) ammonium persulfate and 10 µl TEMED just prior to use.

2 x Sample buffer

25 ml 10% (w/v) SDS, 10 ml glycerol, made up to 50 ml with stacking buffer and

several grains of bromophenol blue added. Prior to use, add 20% (v/v) β-

mercaptoethanol.

Western Blotting Solutions

Appendix 1

160

Transfer Buffer

2.93 g/L glycine, 5.81 g/L Tris base,0.375 g/L SDS, 200 ml/L methanol. Vacuum

filtered

10 x Ponceau stain

20 g/L Ponceau S, 300 g/L trichloroacetic acid, 300 g/L sulfosalicylic acid. Dilute 1 in

10 with ddH2O for use as 1 x Ponceau stain.

10 x TBS

90 g/L NaCL, 12.11 g/L Tris. Adjust pH to 7.4 using HCL and sterilize by autoclaving.

1 x TBS- Tween

100 ml/L 10 x TBS, 1 ml/L Tween® 20.

Blue Native PAGE Solutions

5 x Cathode Buffer

44.8 g/L Tricine, 15.7 g/L Bis-Tris, 1g/L Coomassie 250G. Adjust pH to 7.0 with HCL

at 4ºC and dilute to 1x working solution.

6 x Anode Buffer

62.75 g/L Bis-Tris. Adjust pH to 7.0 with HCL at 4ºC and dilute to 1x working solution.

6x Blue Native Gel buffer

168.8 g/L Amino caprioc acid, 31.2 g/L Bis- Tris. Adjust pH to 7.0 with HCL at 4ºC and

dilute to 1x working solution.

Amino Caprioc Acid (ACA) 750mM

98.4 g/L ACA, 10.46 g/L Bis-Tris, 0.2 g/L EDTA. Adjust pH to 7.0 with HCL at 4ºC.

Appendix 1

161

5% Serva Blue G

0.984 g/L ACA, 0.5 g/L Coomassie 250G.

1 x Sample Buffer

750 µl/ml ACA 750, 150 µl/ml n-Dodecylmaltoside 10%, 150 µl/ml 5% Serva Blue G.

Arabidopsis Mitochondrial isolation media

Grinding media

0.45 M mannitol, 50 mM anhydrous sodium pyrophosphate (10.H2O), 0.5% (w/v) BSA,

0.5% (w/v) PVP-40, 2MM EDTA. Adjust pH to 8.0 using H3PO4. Add 20mM cysteine

just prior to use.

1 x Wash medium

0.3 M mannitol, 10mM TES, 0.2% (w/v) BSA. Adjust pH to 7.5 using HCL.

1st Gradient

0.6 M mannitol, 20mM TES, 0.2% (w/v) BSA. Adjust pH to 7.5 using HCL.

2nd Gradient

0.6 M sucrose, 20mM TES, 0.2% (w/v) BSA. Adjust pH to 7.5 using HCL.

40% (v/v) Gradient 1

40% Percoll® in 50% (v/v) gradient buffer 1 in ddH2O.

21% (v/v) Gradient 1

21% Percoll® in 50% (v/v) gradient buffer 1 in ddH2O.

16% (v/v) Gradient 1

16% Percoll® in 50% (v/v) gradient buffer 1 in ddH2O.

Appendix 1

162

Gradient

5 ml 40% (v/v) gradient 1, 20 ml 21% (v/v) gradient 1, 10 ml 16% (v/v) gradient 1.

In vitro import Solutions

In vitro transcription/translation reaction master mix (50 µl)

25 µl TNT® rabbit reticulocyte lysate, 4µl TNT® reaction buffer, 2 µl T7 or SP6 RNA

polymerase, 2 µl 1 mM amino acid mixture minus methionine, 2 µl RNasin®

ribonuclease inhibitor (40 units/µl; Roche, Australia), 1 µl [35S]-methionine (> 1000

Ci/mmol at 10 mCi/ml; Amersham Pharmacia Biotech, Australia)

2 x Mitochondrial import buffer

0.6 M sucrose, 100 mM KCL, 20 mM Mops, 10 mM KH2PO4, 0.2% (w/v) BSA. Adjust

pH to 7.5 with HCL.

Import Master Mix

1 ml of 2 x import buffer, 2 µl 1 M MgCl2, 20 µl of 100 mM methionine, 4 µl 100 mM

ADP, 15 µl 100 mM ATP, 20 µl 0.5 M succinate, 20 µl 0.5 M DTT. This solution was

made up with ddH2O to 2 ml prior to use.

159

Appendix 2

QRT-PCR Primers. UPOX1, UPOX2 and UPOX3, their chromosomal locus, the primer sets used, their melting temperatures and the optimum primer

concentration given in µM for the respective primer set

Name Accession # Sequence 5’-3’ G/C: A/T Melting

Temperature

Optimum Primer Concentration for

qRT-PCR

UPOX1 Fwd At2g21640 CCGAGAACCCGCCAAAACC 12:7 62 °C 0.9 M

UPOX1 Rev At2g21640 GCTTCTCTGCAACTGCCTC 11:8 60 °C 0.9 M

UPOX2 Fwd At4g39235 GAGACCCAGACAAATCAAAG 9:11 58 °C 0.7 M

UPOX2 Rev At4g39235 CTGCGACCGCCTTTGACAATCC 13:9 70 °C 0.7 M

UPOX3 Fwd At3g05570 GACGCAGAAAACTCAAG 8:9 50 °C 0.7 M

UPOX3 Rev At3g05570 CAGCCTTTGACAACCCAAAC 10:10 60 °C 0.7 M

159

SALK 010761 T-DNA screening primers.

Name Accession # Sequence 5’-3’ CG% Melting Temperature

SALK 010761 Fwd At2g21640 GCTTCGATTCAGTATCACGGG 52 57.4 °C

SALK 010761 Rev At2g21640 CTTAAGGGAGGAAGAGTCATGC 50 56.8 °C

160