10
Immunomodulation of TGF-beta1 in mdx mouse inhibits connective tissue proliferation in diaphragm but increases inflammatory response: Implications for antifibrotic therapy Francesca Andreetta, Pia Bernasconi, Fulvio Baggi, Paolo Ferro, Laura Oliva, Elisa Arnoldi, Ferdinando Cornelio, Renato Mantegazza, Paolo Confalonieri * Department of Neuroimmunology and Neuromuscular Diseases, National Neurological Institute ‘‘Carlo Besta’’, via Celoria 11, 20133 Milan, Italy Received 4 November 2005; received in revised form 14 February 2006; accepted 6 March 2006 Abstract Irreversible connective tissue proliferation in muscle is a pathological hallmark of Duchenne muscular dystrophy (DMD), a genetic degenerative muscle disease due to lack of the sarcolemmal protein dystrophin. Focal release of transforming growth factor-beta1 (TGF-h1) is involved in fibrosis development. Murine muscular dystrophy (mdx) is genetically homologous to DMD and histopathological alterations comparable to those in DMD muscles occur in diaphragm of older mdx mice. To investigate the early development of fibrosis and TGF-h1 involvement, we assessed diaphragms in 6 – 36-week-old mdx and C57/BL6 (control) mice for fibrosis, and used real-time PCR and ELISA to determine TGF-h1 expression. Significantly greater fibrosis and TGF-h1 expression were found in mdx from the 6th week. Mice treated with neutralizing antibody against TGF-h1 had lower levels of TGF-h1 protein, reduced fibrosis, unchanged muscles fiber degeneration/ regeneration, but increased inflammatory cells (CD4 + lymphocytes). These data demonstrate early and progressive fibrosis in mdx diaphragm accompanied by TGF-h1 upregulation. Reduction of TGF-h1 appears promising as a therapeutic approach to muscle fibrosis, but further studies are required to evaluate long term effects of TGF-h1 immunomodulation on the immune system. D 2006 Elsevier B.V. All rights reserved. Keywords: Muscular dystrophy; mdx animal model; Muscle fibrosis; Transforming growth factor-h1; Fibrogenic cytokine; Immunomodulation 1. Introduction Abnormal connective tissue proliferation following myofiber degeneration is a major pathologic feature of Duchenne muscular dystrophy (DMD), a severe genetic myopathy due to a lack of the sarcolemmal protein dystrophin, and clinically characterized by progressive and irreversible degeneration of muscle tissue (Sanes, 1994; Engel et al., 1994). The proliferation of muscle extracellular matrix, characterized by deposition of fibronectin and type I and III collagens in the endomysium and perimysium of muscle tissue (Foidart et al., 1981; Stephens et al., 1982; Duance et al., 1980), leads to irreversible derangement of muscle organization, by impeding the regeneration of muscle fibers and hindering nutritional support, particularly in advanced stages when fibers are physically isolated from their blood supply (Engel et al., 1994; Duance et al., 1980). Since this fibrotic proliferation is likely to be a major obstacle to the efficacy of therapies for muscular dystro- phies, early interventions to prevent it will probably be necessary as part of an effective treatment protocol. Abnormal connective tissue proliferation also occurs in liver cirrhosis, glomerulonephritis, idiopathic lung fibrosis and systemic sclerosis. In these conditions, focal release of fibrogenic cytokines, particularly transforming growth fac- tor-beta1 (TGF-h1) is a key element in promoting fibroblast proliferation and collagen synthesis (Kovacs, 1991). TGF-h1 is a multifunctional cytokine with roles in inflammation, immunomodulation, and wound healing, as well as fibrosis (Border and Noble, 1994). A significant correlation between 0165-5728/$ - see front matter D 2006 Elsevier B.V. All rights reserved. doi:10.1016/j.jneuroim.2006.03.005 * Corresponding author. Tel.: +39 02 23942255; fax: +39 02 70633874. E-mail address: [email protected] (P. Confalonieri). Journal of Neuroimmunology 175 (2006) 77 – 86 www.elsevier.com/locate/jneuroim

Immunomodulation of TGF-beta1 in mdx mouse inhibits connective tissue proliferation in diaphragm but increases inflammatory response: Implications for antifibrotic therapy

Embed Size (px)

Citation preview

www.elsevier.com/locate/jneuroim

Journal of Neuroimmunolo

Immunomodulation of TGF-beta1 in mdx mouse inhibits connective tissue

proliferation in diaphragm but increases inflammatory response:

Implications for antifibrotic therapy

Francesca Andreetta, Pia Bernasconi, Fulvio Baggi, Paolo Ferro, Laura Oliva, Elisa Arnoldi,

Ferdinando Cornelio, Renato Mantegazza, Paolo Confalonieri *

Department of Neuroimmunology and Neuromuscular Diseases, National Neurological Institute ‘‘Carlo Besta’’, via Celoria 11, 20133 Milan, Italy

Received 4 November 2005; received in revised form 14 February 2006; accepted 6 March 2006

Abstract

Irreversible connective tissue proliferation in muscle is a pathological hallmark of Duchenne muscular dystrophy (DMD), a genetic

degenerative muscle disease due to lack of the sarcolemmal protein dystrophin. Focal release of transforming growth factor-beta1 (TGF-h1)is involved in fibrosis development. Murine muscular dystrophy (mdx) is genetically homologous to DMD and histopathological alterations

comparable to those in DMD muscles occur in diaphragm of older mdx mice. To investigate the early development of fibrosis and TGF-h1involvement, we assessed diaphragms in 6–36-week-old mdx and C57/BL6 (control) mice for fibrosis, and used real-time PCR and ELISA

to determine TGF-h1 expression. Significantly greater fibrosis and TGF-h1 expression were found in mdx from the 6th week. Mice treated

with neutralizing antibody against TGF-h1 had lower levels of TGF-h1 protein, reduced fibrosis, unchanged muscles fiber degeneration/

regeneration, but increased inflammatory cells (CD4+lymphocytes). These data demonstrate early and progressive fibrosis in mdx

diaphragm accompanied by TGF-h1 upregulation. Reduction of TGF-h1 appears promising as a therapeutic approach to muscle fibrosis, but

further studies are required to evaluate long term effects of TGF-h1 immunomodulation on the immune system.

D 2006 Elsevier B.V. All rights reserved.

Keywords: Muscular dystrophy; mdx animal model; Muscle fibrosis; Transforming growth factor-h1; Fibrogenic cytokine; Immunomodulation

1. Introduction

Abnormal connective tissue proliferation following

myofiber degeneration is a major pathologic feature of

Duchenne muscular dystrophy (DMD), a severe genetic

myopathy due to a lack of the sarcolemmal protein

dystrophin, and clinically characterized by progressive and

irreversible degeneration of muscle tissue (Sanes, 1994;

Engel et al., 1994). The proliferation of muscle extracellular

matrix, characterized by deposition of fibronectin and type I

and III collagens in the endomysium and perimysium of

muscle tissue (Foidart et al., 1981; Stephens et al., 1982;

Duance et al., 1980), leads to irreversible derangement of

0165-5728/$ - see front matter D 2006 Elsevier B.V. All rights reserved.

doi:10.1016/j.jneuroim.2006.03.005

* Corresponding author. Tel.: +39 02 23942255; fax: +39 02 70633874.

E-mail address: [email protected] (P. Confalonieri).

muscle organization, by impeding the regeneration of

muscle fibers and hindering nutritional support, particularly

in advanced stages when fibers are physically isolated from

their blood supply (Engel et al., 1994; Duance et al., 1980).

Since this fibrotic proliferation is likely to be a major

obstacle to the efficacy of therapies for muscular dystro-

phies, early interventions to prevent it will probably be

necessary as part of an effective treatment protocol.

Abnormal connective tissue proliferation also occurs in

liver cirrhosis, glomerulonephritis, idiopathic lung fibrosis

and systemic sclerosis. In these conditions, focal release of

fibrogenic cytokines, particularly transforming growth fac-

tor-beta1 (TGF-h1) is a key element in promoting fibroblast

proliferation and collagen synthesis (Kovacs, 1991). TGF-h1is a multifunctional cytokine with roles in inflammation,

immunomodulation, and wound healing, as well as fibrosis

(Border and Noble, 1994). A significant correlation between

gy 175 (2006) 77 – 86

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–8678

fibrosis and TGF-h1 expression in Duchenne and Becker

muscular dystrophies has been reported, supporting a role for

this cytokine in the development of muscle fibrosis, and

suggesting it as target for antifibrotic therapies (Bernasconi et

al., 1995; Yamazaki et al., 1994).

Murine X-linked muscular dystrophy (mdx) and DMD are

genetically homologous conditions characterized by a com-

plete absence of dystrophin due to mutations in the

dystrophin gene. The mdx mouse is a useful animal model

for DMD (Hoffman and Dressman, 2001). Although the limb

muscles of adult mdxmice showmuch less weakness, muscle

degeneration and fibrosis than DMD boys, the diaphragm

exhibits severe degeneration and functional impairment

similar to that seen in DMD (Stedman et al., 1991). In fact,

the extracellular matrix of mdx diaphragm muscle doubles

between 60 and 120 days, and is increased by 8-fold at 240

days (Louboutin et al., 1993). However, the extent of fibrosis

and TGF-h1 expression in early stages of the disease is

poorly documented. Hartel et al. (2001) found increased

TGF-h1 expression by ELISA in the diaphragm at 12-week-

old mdx mice, and suggested its involvement in fibrosis;

while Gosselin et al. (2004) reported early overexpression of

TGF-h1 transcripts in mdx diaphragm, as well as inhibition

of type I collagen mRNA, after decorin administration, but

did not assess muscle fibrosis or TGF-h1 protein expression.Studies on animal models of other diseases characterized

by fibrosis have shown that reduction of TGF-h1 levels in

affected tissues (skin, lung, thyroid and kidney) can limit

fibrotic development (McCormick et al., 1999; Chen et al.,

2002; Ziyadeh et al., 2000). To investigate whether the mdx

mouse diaphragm exhibits a pattern of early fibrosis and

early TGF-h1 overexpression, similar to that in human

dystrophic muscle, and to provide indications as to the

utility of early antifibrotic therapy, we studied mdx and C57/

BL6 mice from 6 to 36 weeks of age, assessing fibrosis by

morphometry (De Luca et al., 2005) and using ELISA and

real-time PCR to determine TGF-h1 expression. Since we

found significantly greater fibrosis and TGF-h1 from 6

weeks, we performed additional experiments to test the

effect of limiting TGF-h1 activity by administering mono-

clonal antibody against the cytokine. In view of indications

that TGF-h1 can have both pro- or antiinflammatory effects

(Wahl, 1992), that inflammation can worsen muscle

degeneration (Chen et al., 2000; Porter et al., 2002) and

may interfere with muscle regeneration (Engel and Arahata,

1986; Spencer et al., 2001), we also investigated effects of

anti-TGF-h1 antibody administration on inflammation,

regeneration and degeneration in mdx diaphragm muscle.

2. Materials and methods

2.1. Animals

All experiments were conducted in accordance with

the Italian Guidelines for the use of laboratory animals,

which conform to European Community Directive 86/

609/EEC. Twenty-five mdx mice (Jackson Laboratories,

Bar Harbor, ME, USA) and 25 C57/BL6 mice (Charles

River, Calco, Italy) were used to investigate the

development of muscle fibrosis and expression of TGF-

h1 between the ages of 6 and 36 weeks. Eighteen

additional mdx mice were used to investigate the effects

of anti-TGF-h1 treatment on muscle fibrosis, TGF-h1expression, degeneration, regeneration and inflammation

at 12 weeks. The animals were sacrificed by cervical

dislocation; the diaphragms were removed rapidly, folded,

rolled up and frozen in isopentane pre-cooled in liquid

nitrogen.

2.2. Anti-TGF-b1 treatment

Eleven mdx mice of age 6 weeks were injected

intraperitoneally with 300 Ag of anti-TGF-h1(1D11.16.8, HB 9849 ATCC, Manassas, VA, USA) on

alternate days to age 12 weeks. Seven mdx mice of the

same age were injected intraperitoneally at the same times

with 300 Ag normal mouse IgG (Pierce, Rockford, IL,

USA). During anti-TGF-h1 treatment, the mdx mice did

not shown abnormal behaviour, or differences in gross

vital functions compared to untreated mdx mice and IgG-

treated mice.

2.3. Morphometric analysis

Morphometric analysis was carried out on C57/BL6

mice as controls and also on untreated mdx mice and on

those treated with anti-TGF-h1 or normal IgG. Serial

cryostat cross-sections (6–8 Am thick) of rolled dia-

phragm were stained with Meyer’s haematoxylin and

eosin or Masson trichrome. The extent of endomysial and

total (endomysial plus perimysial) connective tissue was

determined on haematoxylin and eosin-stained sections

(with Masson trichrome-stained sections used to check

morphology) at �20 magnification using the NIH Image

software. At least three randomly selected fields from each

section were analyzed. The area of connective tissue as a

percentage of total muscle in each field was calculated, and

the mean percentage for each group of animals calculated (De

Luca et al., 2005).

2.4. Immunostaining

Immunohistochemical analysis was carried out on

untreated mdx mice and on those treated with anti-

TGF-h1 or normal IgG. Serial 6–8-Am-thick cryostat

cross-sections of rolled diaphragm were cut, collected

onto polylysine-coated slides, and fixed with ice-cold

acetone for 1 min. Subsequent steps were performed in a

humid chamber at room temperature. The sections were

treated with peroxidase block solution (DAKO, Glostrup,

Denmark) for 5 min followed by protein block solution

0%

5%

10%

15%

20%

25%

30%

6w 12w 18w 24w 36wage

*A

0%

5%

10%

15%

20%

25%

30%

6w 12w 18w 24w 36wage

*

B

% o

f end

omys

ial c

onne

ctiv

e tis

sue

% o

f to

tal c

onne

ctiv

e tis

sue

Fig. 1. Histograms showing development of total (A) and endomysial (B)

fibrosis. Mdx mice are characterized by increasing fibrosis with age, with a

significant increase between 6 and 12 weeks (*P�0.001, Student’s t-test).

Open bars: C57/BL6 controls; black bars: mdx mice.

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–86 79

(DAKO) for 10 min. The sections were then incubated

for 90 min with primary antibody. The following

antibodies were used: anti-NCAM (Chemicon, Intern,

Germany) a marker of regenerating fibers (Spencer and

Mellgren, 2002); anti-CD11b (BD Pharmingen, San

Fig. 2. Digital images of hematoxylin and eosin-stained diaphragm cross-sections.

weeks. In (A) note the normal organization of muscle tissue, with fibers of similar

the progressive disorganization of fascicle architecture with abnormal variation in

arrowheads indicate expanding endomysial fibrosis. Scale bar: 60 Am.

Diego, CA, USA) marker of degeneration staining

macrophages and neutrophils (Spencer et al., 2000);

anti-CD4 (helper T-lymphocytes) (BD Pharmingen); and

anti-CD8a (suppressor T-lymphocytes) (Cederlane,

Hornby, Ontario, Canada).

Sections were then washed in PBS and incubated with

anti-rat IgG biotinylated secondary antibody (Jackson

ImmunoResearch Laboratories, West Grove, PA, USA)

for 1 hour, followed by avidin biotin horseradish perox-

idase complex (ABC kit, Vector Laboratories Inc, Burlin-

game, CA, USA) for 30 min. The chromogen was DAB

(DAKO) applied for up to 10 min followed by brief (1

min) counterstaining with Meyer’s haematoxylin.

Sections were examined under a Zeiss Axiophot micro-

scope and digital images were obtained with a Leica DC-

500 Firecam digital camera.

2.5. Assessment of regeneration, degeneration and

inflammation

These analyses were carried out on untreated mdx

mice, and on those treated with anti-TGF-h1 or normal

IgG. Consecutive frozen cross-sections of rolled dia-

phragm stained to reveal NCAM and CD11b, were

assessed at �20 magnification using NIH Image soft-

ware. At least three randomly selected fields from each

section were analyzed. Regenerating and degenerating

areas were expressed, respectively, as the percentages of

NCAM- and CD11b-positive areas of total muscle in each

field.

To assess inflammation, the total numbers of CD4+ and

CD8+ lymphocytes were counted on entire cross sections

of rolled diaphragm, and normalised per unit area.

(A) C57/BL6 mice at 12 weeks; (B–F) mdx mice at 6, 12, 18, 24, and 36

size and only thin strands (arrowheads) of connective tissue. In (B–F) note

fiber size (double arrow) and replacement by fibrous connective tissue. The

00,5

11,5

22,5

33,5

4

6w 12w 18w 24w 36wage

*

*A

0

1

2

3

4

5

6

7

8

6w 12w 18w 24w 36wage

B*

TG

F-β1

mR

NA

rela

tive

valu

e(p

g T

GF-

β1/ μ

g to

t. pr

ot.)

TG

F-β1

Fig. 3. Quantitation of TGF-h1 protein (ELISA) and transcripts (RT-PCR)

in diaphragm muscle of mdx and C57/BL6 mice of ages 6–36 weeks. (A)

TGF-h1 protein is significantly upregulated at 12 weeks and 24 weeks

(*P�0.001, Student’s t-test). Open bars C57/BL6 controls; black bars mdx

mice. (B) TGF-h1 transcript is overexpressed in mdx diaphragm only at 24

weeks of age (*P=0.0051, Student’s t-test). Open squares: C57/BL6 mice;

black triangles: mdx mice.

0%

5%

10%

15%

20%

25%

30%

% o

f to

tal c

onne

ctiv

e tis

sue

*

C57/BL6 mdx + IgG + anti-TGF-β1

A

*

0%

5%

10%

15%

20%

25%

% o

f en

dom

ysia

l con

nect

ive

tissu

e

*

C57/BL6 + IgGmdx + anti-TGF-β1

B

*

Fig. 4. Effect of treatment with antibody to TGF-h1 from 6 to 12 weeks

on extent of muscle connective tissue. Histograms showing total (A) and

endomysial (B) connective tissue in 12-week-old animals. Levels of total

and endomysial connective tissue are significantly reduced in mdx mice

treated with anti-TGF-h1 antibody compared to untreated and IgG-treated

mice (*P�0.001, Kruskal–Wallis test). In these mice the amount of

fibrosis was similar to that found in untreated control (C57/BL6) mice.

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–8680

2.6. Determination of TGF-b1 protein

TGF-h1 protein was quantitated by ELISA, according

to the manufacturer’s instructions (R&D System, Minne-

apolis, MN, USA), on diaphragms from C57/BL6 mice,

untreated mdx mice, anti-TGF-h1-treated mdx mice and

IgG-treated mdx mice. Briefly, 10–20 mg of diaphragm

was homogenized in 500 Al of a solution containing 1%

Triton X-100, 20 mM Tris pH 8.0, 137 mM sodium

chloride, 10% glycerol, 5 mM ethylendiaminetetraacetic

acid and 1 mM phenylmethylsulphonyl fluoride and

treated as described (Hartel et al., 2001). TGF-h1 levels

were expressed as pg of TGF-h1/Ag of total protein.

2.7. Determination of TGF-b1 transcript

Total RNA was extracted from 10 to 20 mg of mdx and

age-matched C57/BL6 diaphragms using RNAwiz reagent

(Ambion, Woodward Austin, TX, USA). The extract was

treated with DNase I (Ambion). Random-primed cDNAwas

prepared using Superscript II reverse transcriptase (Invitro-

gen, Carlsbad, CA, USA) following the manufacturer’s

instructions and stored at �20 -C pending amplification.

For quantitation of TGF-h1, predesigned functionally

tested assay (Mm00441724_m1, Applied Biosystems, Foster

City, CA, USA) was used. cDNA samples (each

corresponding to 100 ng total RNA) were amplified in

triplicate using a GeneAmp 5700 Sequence Detection System

(Applied Biosystems) in a volume of 25 Al containing

TaqMan Universal PCR Master Mix (containing AmpliTaq

Gold DNA polymerase), 1 Al of predesigned TGF-h1 primers

and probes. 18S rRNA (Applied Biosystems) was used as

endogenous control. The appearance of specific fluorescence

from TGF-h1 and 18S mRNAwas analyzed using Sequence

Detector Software (version 1.6, Applied Biosystems). The

expression of TGF-h1 in each diaphragm sample was

normalized to that of 18S and calculated from the formula

2�DDCT as described in the manufacturer’s instructions

(user bulletin #2, Applied Biosystems). As calibrator for

untreated mdx and C57/BL6 mice, the normalized TGF-h1CT value from diaphragm of 6-week-old C57/BL6 mice

was used; for anti-TGF-h1- and IgG-treated mice the mean

normalized CT value for diaphragms from 12-week-old

C57/BL6 mice was used.

2.8. Statistical analysis

Data are expressed as means and standard deviations

(TS.D.). StatView software, release 5.01 (SAS Institute

Fig. 5. Digital images of Masson’s trichrome-stained diaphragm cross-sections from 12-week-old mice. (A) Untreated mdx mice; (B) mdx mice treated

with mouse IgG; (C) mdx mice treated with anti-TGF-h1 antibody. Masson’s trichrome allows good visualization of extracellular matrix and connective

tissue. Note the disorganization of muscle tissue structure in (A) and (B) due to fibrosis (square), and good preservation of syncytial structure in (C)

(square). The arrows indicate endomysial fibrosis which is more evident in untreated and IgG-treated mice than anti-TGF-h1-treated mice. Scale bar:

60 Am.

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–86 81

Inc., Cary, North Carolina, USA) and Prism software

(GraphPad software, San Diego, California, USA) were

used for the statistical analyses. The two-tailed Student’s

t-test with Bonferroni correction and Kruskal–Wallis with

Dunn’s multiple comparison test were used to assess the

differences between C57 control mice and mdx mice and

between experimental and control groups. P values �0.05were considered significant.

Table 1

Effect of the anti-TGF-h1 antibody and normal IgG treatment on extent of fibrosis,

numbers of CD4 and CD8 cells per unit area

Mice % of total

fibrosis

% of endomysial

fibrosis

mdx 1 20.53T3.64 15.21T2.282 23.24T1.96 17.78T2.93

3 23.97T1.05 17.70T2.63

4 26.07T0.49 19.29T3.09

5 22.42T1.98 17.14T3.47MeanTS.D. 23.25T2.60 17.42T2.81

mdx+IgG 1 14.86T2.06 11.60T0.89

2 18.81T2.33 13.27T0.71

3 16.28T0.66 12.76T1.10

4 21.65T2.25 18.13T3.165 18.99T1.15 14.65T1.71

6 19.59T2.58 15.51T1.35

7 20.36T0.97 15.76T2.02MeanTS.D. 18.65T2.71 14.53T2.54

mdx+anti-TGF-h1 1 11.68T1.15 9.84T0.70

2 12.50T1.15 10.01T1.38

3 13.34T2.82 9.86T1.18

4 9.24T1.04 7.82T0.665 9.71T1.38 8.51T0.59

6 12.29T1.53 9.84T1.95

7 10.74T0.66 9.62T0.77

8 10.48T1.64 9.92T0.689 12.67T0.58 11.77T1.07

10 10.65T0.81 8.59T1.64

11 12.33T1.36 10.18T1.14MeanTS.D. 11.36T1.66 9.63T1.40

MeansTS.D. for each animal and for each group are shown. The percentages of tot

The data for regeneration and degeneration are more variable, but differences are n

significantly between the three groups and was higher in mdx mice treated with an

CD8 cells did not differ significantly between the three groups (see Fig. 9).

3. Results

3.1. Fibrosis and cytokine expression with age

In diaphragms of mdx mice, the quantity of total and

endomysial connective tissue as a proportion of total

muscle increased significantly from the youngest age

examined (6 weeks) to 12 weeks (P�0.001, Student’s

percentages of regenerating and degenerating cell aspects per unit areas and

% of

regeneration

% of

degeneration

CD4

(mm2)

CD8

(mm2)

5.53T1.75 5.30T0.74 11 3

6.34T3.58 4.60T1.38 9 2

3.65T1.09 3.74T0.62 6 0

7.04T3.69 5.04T1.52 5 1

7.65T5.71 3.91T2.13 4 0

6.04T3.47 4.52T1.35 7T2.9 1T0.6

2.41T0.01 1.22T0.02 6 2

10.07T9.39 2.55T0.49 11 1

6.74T0.02 2.79T0.11 5 1

11.70T0.14 2.63T1.12 1 0

5.84T2.36 2.59T0.80 1 0

4.42T0.36 3.51T2.64 12 0

5.66T4.94 2.98T1.39 1 0

6.69T4.60 2.61T1.43 5T4.7 1T0.7

3.61T0.23 4.09T1.96 16 5

16.46T6.63 4.77T2.49 10 1

6.11T3.42 2.78T0.83 24 2

18.52T21.17 3.93T0.41 10 1

12.33T13.85 2.62T1.30 11 2

4.94T0.10 2.66T0.21 9 1

5.00T1.30 4.45T0.94 22 1

2.81T0.32 5.12T0.12 40 9

9.48T0.37 6.66 T1.88 21 6

12.15T13.43 4.89T1.65 16 2

2.81T1.04 2.68T1.68 33 13

8.57T8.60 4.06T1.73 19T10.1 4T3.9

al and endomysial fibrosis are homogeneous within each group (see Fig. 4).

ot significant (see Fig. 8). The number of CD4 T cells per unit area differed

ti-TGF-h1 antibody than untreated and IgG treated animals. The number of

00.10.20.30.40.50.60.70.80.9

TG

F-β1

(pg

TG

F-β1

/μg

tot.

prot

.)

*

C57/BL6 mdx + anti-TGF-β1+ IgG

A**

00.20.40.60.8

11.21.41.61.8

2

TG

F-β1

mR

NA

rel

ativ

e va

lue

C57/BL6 + IgGmdx + anti-TGF-β1

B **

Fig. 6. Quantitation of TGF-h1 protein (ELISA) and transcripts (RT-PCR)

in homogenized diaphragm from 12-week-old mice. (A) Mdx mice

treated with anti-TGF-h1 antibody had significantly lower protein levels

than untreated (*P�0.01, Kruskal–Wallis test) and IgG-treated mdx

mice (**P�0.05, Kruskal–Wallis test). (B) Mice treated with anti-TGF-

h1 antibody and with normal IgG had significantly lower transcript

levels of TGF-h1 than untreated mdx mice (*P�0.05, Kruskal–Wallis

test).

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–8682

t-test). Thereafter the extent of total and endomysial

connective tissue remained high and at levels close to

those observed at 12 weeks (Figs. 1 and 2). The extents of

total and endomysial connective tissue were significantly

greater (P�0.001, Student’s t-test) in mdx mice than

C57/BL6 mice at 6 weeks, and in all the older ages

(Figs. 1 and 2).

3.2. TGF-b1 protein in mouse diaphragm with increasing

age

The quantity of TGF-h1 in the diaphragm of C57/BL6

mice did not change significantly with age (Fig. 3A). The

quantity of TGF-h1 protein was significantly greater in mdx

diaphragms than C57/BL6 mice at 12 weeks (0.610T0.102vs. 0.236T0.071; P�0.001, Student’s t-test), and 24 weeks

(2.516T0.982 vs. 0.317T0.029; P�0.001, Student’s t-test)

(Fig. 3A). Differences were not significant at 6, 18 and 36

weeks.

3.3. TGF-b1 transcript in mouse diaphragm with increasing

age

TGF-h1 transcript levels were quantified by real-time

PCR in mdx and C57/BL6 mouse diaphragms taken at

different ages (from 6 to 36 weeks of age; with an average

of three animals per group investigated); as calibrator the

normalized CT mean TGF-h1 value (17.26) in 6-week-old

C57/BL6 mice was used. In C57/BL6 mice TGF-h1 was

transcribed at constant rate at all ages (Fig. 3B). In mdx

mice TGF-h1 transcript levels at 6, 12 and 18 weeks

were similar (TGF-h1 relative value: 0.873T0.196; 1.042T0.597; 0.958T0.237) compared to C57/BL6 mice; at 24

weeks, levels were 6-fold greater than C57/BL6 of the same

age (P=0.0051, Student’s t-test) and declined thereafter

(Fig. 3B).

3.4. Effects of treatments with anti-TGF-b1 antibody and

normal mouse IgG

3.4.1. Morphometric evaluation of connective tissue

Twelve-week-old mdx mice treated for 6 weeks with

anti-TGF-h1 antibody had significantly less total and

endomysial connective tissue than untreated mdx mice

(P�0.001, Kruskal–Wallis test). Mdx mice treated with

normal mouse IgG did not differ in terms of extent of

connective tissue from untreated mdx mice, but differed

significantly from mice treated with anti-TGF-h1 antibody

(P�0.001, Kruskal–Wallis test) (Figs. 4 and 5, Table 1).

Note that treatment with anti-TGF-h1 antibody brought

levels of connective tissue close to those of untreated

C57/BL6 control mice (Fig. 4).

3.4.2. TGF-b1 protein and transcript

Twelve-week-old mdx mice treated for 6 weeks with

anti-TGF-h1 antibody had significantly less (P�0.01,

Kruskal–Wallis test) TGF-h1 protein in diaphragm than

untreated mdx mice (0.247T0.118 vs. 0.610T0.102) or

mice treated with normal IgG (0.522T0.295: P <0.05,

Kruskal–Wallis test). Protein levels in anti-TGF-h1-treatedmice were similar to those in diaphragm from C57/BL6

control mice (0.200T0.073) (Fig. 6A).Levels of TGF-h1 transcript were significantly lowered

by both anti-TGF-h1 antibody and normal IgG treatments

(0.605T0.155, P�0.05, and 0.666T0.403, P�0.05, Krus-

kal–Wallis test) (Fig. 6B).

3.4.3. Muscle regeneration, degeneration and inflammatory

cell content

Twelve-week-old mdx mice diaphragms contained

NCAM-positive regenerating and CD11b-positive degener-

ating fibers. Each fiber type was usually present in clumps.

Sometimes regenerating and degenerating fibers were

localised close to each other (Fig. 7B, E and C, F).

Quantitation of regenerating and degenerating areas did not

reveal significant differences between mdx mice treated

and not treated with anti-TGF-h1 antibody (Fig. 8 and

Table 1).

0%

5%

10%

15%

20%

% o

f re

gene

ratio

n an

dde

gene

ratio

n ar

eas

mdx + anti-TGF-β1+ IgG

Fig. 8. Evaluation of muscle fiber regeneration/degeneration in mdx

untreated and treated mice. Treatment with normal mouse IgG or with

antibody against TGF-h1 did not cause significant changes in the extent of

regeneration (NCAM staining=open bars) or degeneration (CD11b stai-

ning=black bars).

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–86 83

Twelve-week-old untreated and IgG-treated mdx mice

had low numbers of CD4+ and CD8+ T lymphocytes

scattered in perimysial and endomysial connective tissue.

These lymphocytes were never found in groups of more

than three cells and never appeared to be invading muscle

fibers (Fig. 7G, J and H, K). Mdx mice treated with anti-

TGF-h1 antibody have significantly more CD4+ T cells per

unit area of muscle than untreated (P <0.05, Kruskal–

Wallis test) or normal IgG-treated ( p <0.01, Kruskal–Wallis

test) mdx mice. These cells were occasionally present in

groups in the endomysium, but again there appeared to be

no invasion of the muscle fibers. The number of CD8+ T

cells was also numerically greater in the diaphragm of

antibody-treated mdx mice without reaching statistical

Fig. 7. Digital images of consecutive diaphragm sections of untreated and IgG- and anti-TGF-h1-treated mdx mice showing staining with NCAM for

regenerating fibers (A–C) (arrows), CD11b for degenerating areas (D–F) (arrows), CD4 (G–I) and CD8 (J–L) (arrows). Note that degeneration, regeneration

and inflammatory cells are often localized in the same areas. In anti-TGF-h1-treated mdx mice an increase of CD4 T cells is evident (I). Scale bar: 60 Am.

0

5

10

15

20

25

30

35

CD

4 an

d C

D8

T-c

ells

/ m

m2

*

mdx + anti-TGF-β1+ IgG

**

Fig. 9. Quantitative evaluation of numbers per unit area of CD4 and CD8

cells in untreated mdx mice and those treated with normal IgG and antibody

against TGF-h1. Treatment with anti-TGF-h1 antibody was associated withsignificantly more CD4 cells than untreated (*P�0.05, Kruskal–Wallis

test) and IgG-treated mice (**P�0.01, Kruskal–Wallis test). Open bars:

CD4 T-cells; black bars: CD8 T-cells.

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–8684

significance; these cells never appeared to invade muscle

fibers (Figs. 7I, L and 9 and Table 1).

4. Discussion

Our first finding is that connective tissue formed a

greater proportion of the diaphragm muscle of mdx mice

(15.5%) than of control C57/BL6 mice (11%) at the

earliest age studied (6 weeks). The difference was

significant at 12 weeks (23% vs. 9%) and reached 25%

at 36 weeks (Fig. 1, Table 1). Thus the fibrotic process

(Fig. 2) begins at a very early stage in mdx diaphragm,

just as it does in the limb and other muscles of DMD boys

(Bernasconi et al., 1995). We found that the fibrosis was

present in perimysial septa surrounding muscle fascicula,

and also in the endomysium surrounding individual muscle

fibers, thereby hindering nutritional support by isolating

the myofibers from the blood supply, as also reported in

DMD (Engel et al., 1994).

Our second main finding is that the fibrogenic cytokine

TGF-h1 was upregulated in young mdx mice diaphragm,

with levels numerically higher than control from 6 weeks, a

significant difference at 12 weeks, and a peak difference at

24 weeks (Fig. 3A). These findings enlarge the data of a

previous study that investigated TGF-h1 in diaphragm from

mdx mice at 12 weeks only, finding upregulation of the

protein at that time (Hartel et al., 2001).

Note, however, that we found significantly increased

transcript levels at 24 weeks only (Fig. 3B). A previous

study reported that TGF-h1 transcript levels were signif-

icantly upregulated in mdx diaphragm at 6 and 9 weeks,

but not 12 weeks, and were apparently related to increases

in collagen type I mRNA (Gosselin et al., 2004). The

difference in the timing of TGF-h1 transcript peaks

between the two studies might be related to the fact that

we used real time PCR, whereas the other study used

competitive PCR. Furthermore, we normalized TGF-h1values to those of 18S transcript to obviate differences due

to quantity of starting material. Despite this discrepancy,

several studies indicate that TGF-h1 expression in mdx

mouse diaphragm changes with time, as it does in DMD

limb muscle (Bernasconi et al., 1995).

In view of the similarity of the pathological changes in

mdx diaphragm and DMD limb muscle, particularly in

terms of early development of fibrosis and concomitant

upregulation of TGF-h1, we administered monoclonal

antibody against TGF-h1 to determine whether it was able

to inhibit the development of fibrosis. Treatment from 6

to 12 weeks of age resulted in much lower levels of the

cytokine protein in the diaphragm of treated than

untreated and IgG-treated mdx mice (Fig. 6A). In

addition, TGF-h1 mRNA was inhibited in both treatments,

probably because of the known immunomodulatory effect

of IgG in muscle, as reported in patients treated with high

dose intravenous immunoglobulin for inflammatory myo-

pathies (Amemiya et al., 2000). More importantly, a

significant reduction of muscle fibrosis compared to

untreated mdx mice and also mdx mice treated with

normal IgG (Fig. 4, Table 1) was found. A similarly

successful immuno-modulatory approach to the in vivo

control of this overexpressed cytokine has been reported

in animal models of diseases involving excessive fibrosis

of skin, lung and thyroid (McCormick et al., 1999; Chen

et al., 2002).

Our finding that TGF-h1 immunomodulation inhibits

fibrosis, together with the data of Gosselin et al. (2004)

indicating that the use of decorin to inhibit the biological

activity of TGF-h1 results in the downregulation of type I

collagen mRNA, constitute additional evidence for the

involvement of TGF-h1 in the development of fibrosis,

but more importantly suggest that TGF-h1 inhibition may

be useful as an early clinical approach to the inhibition of

fibrosis in dystrophic disease.

It is noteworthy that treatment with anti-TGF-h1antibody did not eliminate the cytokine from diaphragm

muscle, but brought it to levels similar to those of healthy

control mice (Fig. 6). This finding could be important in

view of the fact that TGF-h1 is involved in multiple

biological processes (Wahl, 1994), has various, sometimes

opposing effects on immune system cells (Wahl, 1992),

may be produced by various cell types including

lymphocytes and macrophages and also platelets (Barnard

et al., 1990) and could be involved in muscle degeneration

and regeneration of muscle fibers, as shown by the close

relationship between TGF-h1 expression and inflammatory

infiltrates in dystrophic muscle (Gosselin et al., 2004), and

by the known inhibitory effects of this cytokine on myoblast

differentiation (Heino and Massague, 1990).

The continuous fiber regeneration that occurs in mdx

muscle is considered the main reason for the good clinical

outcomes in this animal model, while the transient and

limited muscle regeneration that occurs in DMD muscle is

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–86 85

unable to counteract the continuous fiber loss (Granchelli et

al., 2000). In our mice, the 6 weeks of treatment with anti-

TGF-h1 antibody did not affect muscle regeneration or

degeneration (Fig. 8, Table 1) although numbers of CD4 T

cells in the muscle endomysium were increased. The CD8 T

cells did not show signs of direct cytotoxicity (invasion of

non-necrotic muscle fibers) against muscle fibers. This

inflammatory cell pattern is consistent with data reported by

others in limb muscles of untreated mdx mice (Spencer et

al., 1997).

TGF-h1 is a potent inhibitor of lymphocyte activation

and proliferation (Miyazono, 2000; Wahl et al., 1988). Its

inhibition following skeletal muscle injury results in

macrophage infiltration (Lefaucheur et al., 1996). Further-

more TGF-h1-null mice are characterized by dramatic

inflammation (Kulkarni and Karlsson, 1993). The in-

creased inflammatory cells in the muscle of mice treated

with antibody against TGF-h1 is therefore probably due

to reduction in level of the cytokine (TGF-h1), and could

plausibly result in increased muscle degeneration and

fibrosis, particularly in the prolonged treatment that would

be required in DMD therapy. For these reasons, future

antifibrotic protocols with antibodies against TGF-beta1

might also include immunosuppressive agents. In fact it

has been shown that antibody depletion of CD8 and CD4

cells significantly reduces histologically discernable pa-

thology in mdx muscle (Spencer et al., 2001), and that

mdx mice with nu/nu genotype (T cell-deficient) have

reduced muscle fibrosis (Morrison et al., 2000), although

these data were not confirmed by a recent paper

(Morrison et al., 2005). It seems clear that secondary

chronic inflammation could contribute to disease progres-

sion in DMD patients: indeed this is the rationale for

immunomodulatory therapy with glucocorticoids that is

currently the mainstay of treatment in the disease (Skuk et

al., 2002). It is important therefore that proposed anti-

fibrotic treatments for DMD do not exacerbate this

chronic inflammatory state. Our results support the utility

of systemic immunomodulation to inhibit TGF-h1 in

muscle, and hence fibrosis, and provide further evidence

that TGF-h1 plays an important role in the development

of muscle fibrosis, at least in the murine form of x-linked

muscular dystrophy. TGF-h1 inhibition therefore appears

as a promising therapeutic strategy for fibrosis inhibition

in DMD, particularly since no side effects were observed

in the animals during the 6 weeks of treatment. However

effects of TGF-h1 inhibition must be evaluated over the

long-term, paying attention to effects on muscle degener-

ation, muscle fibrosis, and the immune system. Finally,

since muscle fibrosis is likely to be the result of multiple

mechanisms involving molecules other than TGF-h1, datafrom expression profile studies on laser-microdissected

areas of muscle extracellular matrix may identify other

cytokines or growth factors whose modulation may

prevent or reduce fibrosis development without adversely

affecting the immune system.

Acknowledgements

The authors thank Dr. Marina Mora and Prof. Annamaria

De Luca for the helpful suggestions and Donald Ward for

help with the English.

References

Amemiya, K., Semino-Mora, C., Granger, R.P., Dalakas, M.C., 2000.

Downregulation of TGF-beta1 mRNA and protein in muscles of

patients with inflammatory myopathies after treatment with high-dose

intravenous immunoglobulin. Clin. Immunol. 94, 99–104.

Barnard, J.A., Lyons, R.M., Moses, H.L., 1990. The cell biology of TGF-

beta. Biochem. Biophys. Res. Commun. 163, 56–63.

Bernasconi, P., Torchiana, E., Confalonieri, P., Morandi, L., Brugnoni, R.,

Barresi, R., Mora, M., Cornelio, F., Mantegazza, R., 1995. Expression

of transforming growth factor-beta1 in dystrophic patient muscles

correlates with fibrosis: pathogenetic role of fibrogenic cytokines. J.

Clin. Invest. 96, 1137–1144.

Border, W.A., Noble, N.A., 1994. Transforming growth factor-beta in tissue

fibrosis. N. Engl. J. Med. 331, 1286–1292.

Chen, Y.W., Zhao, P., Borup, R., Hoffman, E.P., 2000. Expression profiling

in the muscular dystrophies: identification of novel aspects of molecular

pathophysiology. J. Cell Biol. 151, 1321–1336.

Chen, K., Wei, Y., Shrap, G.C., Braley-Mullen, H., 2002. Inhibition of

TGFbeta1 by anti-TGFbeta1 antibody or lisinopril reduces thyroid

fibrosis in granulomatous experimental autoimmune thyroiditis. J.

Immunol. 169, 6530–6538.

De Luca, A., Nico, B., Liantonio, A., Di donna, M.P., Fraysse, B., Pierno,

S., Burdi, R., Mangeri, D., Rolland, J.F., Camerino, C., Zallone, A.,

Confalonieri, P., Andreetta, F., Arnoldi, E., Courdier-Fruh, I., Mayar,

J.P., Frigeri, A., Pisoni, M., Svelto, M., Conte-Camerino, D., 2005. A

multidisciplinary evaluation of the effectiveness of Cyclosporine A in

dystrophic mdx mice. Am. J. Pathol. 166, 477–489.

Duance, V.C., Stephens, H.R., Dunn, M., Bailey, A.J., Dubowitz, V., 1980.

A role for collagen in the pathogenesis of muscular dystrophy? Nature

284, 470–472.

Engel, A.G., Arahata, K., 1986. Mononuclear cells in myopathies:

quantitation of functionally distinct subsets, recognition of antigen-

specific cell-mediated cytotoxicity in some diseases. Human Pathol. 17,

704–721.

Engel, A.G., Yamamoto, M., Fischbeck, K.H., 1994. Dystrophinopathies.

In: Engel, A.G., Franzini-Armstrong, C. (Eds.), Myology, vol. 2.

McGraw-Hill, New York, pp. 1133–1187.

Foidart, M., Foidart, J.M., Engel, W.K., 1981. Collagen localization in

normal and fibrotic skeletal muscle. Arch. Neurol. 38, 152–157.

Gosselin, L.E., Williams, J.E., Deering, M., Brazeau, D., Koury, S.,

Martinez, D.A., 2004. Localization and early time course of TGF-beta1

mRNA expression in dystrophic muscle. Muscle Nerve 30, 645–653.

Granchelli, J.A., Pollina, C., Hudecki, M.S., 2000. Pre-clinical screening of

drugs using the mdx mouse. Neuromuscul. Disord. 10, 235–239.

Hartel, J.V., Granchelli, J.A., Hudecki, M.S., Pollina, C.M., Gosselin, L.E.,

2001. Impact of prednisone on TGF-beta1 and collagen in diaphragm

muscle from mdx mice. Muscle Nerve 24, 428–432.

Heino, J., Massague, J., 1990. Cell adhesion to collagen and decreased

myogenic gene expression implicated in the control of myogenesis by

transforming growth factor beta. J. Biol. Chem. 265, 10181–10184.

Hoffman, E.P., Dressman, D., 2001. Molecular pathophysiology and

targeted therapeutics for muscular dystrophy. Trends Pharmacol. Sci.

22, 465–470.

Kovacs, E.J., 1991. Fibrogenic cytokines: role of immune mediators in the

development of scar tissue. Immunol. Today 12, 17–23.

Kulkarni, A.B., Karlsson, S., 1993. Transforming growth factor-beta 1

knockout mice: a mutation in one cytokine causes a dramatic

inflammatory disease. Am. J. Pathol. 143, 3–9.

F. Andreetta et al. / Journal of Neuroimmunology 175 (2006) 77–8686

Lefaucheur, J.P., Gjata, B., Lafont, H., Sebille, A., 1996. Angiogenic and

inflammatory responses following skeletal muscle injury are altered by

immune neutralization of endogenous bFGF, IGF1 and TGF-beta1. J.

Neuroimmunol. 70, 37–44.

Louboutin, J.P., Fichter-Gagnepain, V., Thaon, E., Fardeau, M., 1993.

Morphometric analysis of mdx diaphragm muscle fibers. Comparison

with hindlimb muscles. Neuromuscul. Disord. 3, 463–469.

McCormick, L.L., Zhang, Y., Tootell, E., Gilliam, A.C., 1999. Anti-TGF-

beta treatment prevents skin and lung fibrosis in murine scleroderma-

tous graft-versus-host disease: a model for human scleroderma. J.

Immunol. 163, 5693–5699.

Miyazono, K., 2000. Positive and negative regulation of TGF-beta

signaling. J. Cell. Sci. 113, 1101–1109.

Morrison, J., Lu, Q.L., Pastoret, C., Partridge, T., Bou-Gharios, G., 2000. T-

cell-dependent fibrosis in the mdx dystrophic mouse. Lab. Invest. 80,

881–891.

Morrison, J., Palmer, D.B., Cobbold, S., Partridge, T., Bou-Gharios, G.,

2005. Effects of T-lymphocyte depletion on muscle fibrosis in the mdx

mouse. Am. J. Pathol. 166, 1701–1710.

Porter, J.D., Khanna, S., Kaminski, H.J., Rao, J.S., Merriam, A.P.,

Richmonds, C.R., Leahy, P., Li, J., Guo, W., Andrade, F.H., 2002. A

chronic inflammatory responses dominates in skeletal muscle molecular

signature in dystrophin-deficient mdx mice. Hum. Mol. Genet. 11,

263–272.

Sanes, J.R., 1994. The extracellular matrix. In: Engel, A.G., Franzini-

Armstrong, C. (Eds.), Myology, vol. 1. McGraw-Hill, New York, pp.

242–260.

Skuk, D., Vilquin, J.T., Tremblay, J.P., 2002. Experimental and therapeutic

approach to muscular dystrophies. Curr. Opin. Neurol. 15, 563–569.

Spencer, M.J., Mellgren, R.L., 2002. Overexpression of a calpastatin

transgene in mdx muscle reduces dystrophic pathology. Hum. Mol.

Genet. 11, 2645–2655.

Spencer, M.J., Walsh, C.M., Dorshkind, K.A., Rodriguez, E.M., Tidball,

J.G., 1997. Myonuclear apoptosis in dystrophic mdx muscle occurs by

perforine-mediated cytotoxicity. J. Clin. Invest. 99, 2745–2751.

Spencer, M.J., Marino, M.W., Winckler, W.M., 2000. Altered pathological

progression of diaphragm and quadriceps muscle in TNF-deficient,

dystrophin-deficient mice. Neuromuscul. Disord. 10, 612–619.

Spencer, M.J., Montecino-Rodriguez, E., Dorshkind, K., Tidball, J.G., 2001.

Helper (CD4(+)) and cytotoxic (CD8(+)) T-cells promote the pathology

of dystrophin-deficient muscle. Clin. Immunol. 98, 235–243.

Stedman, H.H., Sweeney, H.L., Shrager, J.B., Maguire, H.C., Panettieri,

R.A., Petrof, B., Narusawa, M., Leferovich, J.M., Sladky, J.T., Kelly,

A.M., 1991. The mdx mouse diaphragm reproduces the degenerative

changes of Duchenne muscular dystrophy. Nature 352, 536–539.

Stephens, H.R., Duance, V.C., Dunn, M.J., Bailey, A.J., Dubowitz, V., 1982.

Collagen types in neuromuscular diseases. J. Neurol. Sci. 53, 45–62.

Wahl, S.M., Hunt, D.A., Wong, H.L., Dougherty, S., McFarney-Francis, N.,

Wahl, L.M., Ellingsworth, L., Schmidt, J.A., Hall, G., Roberts, A.B.,

Sporn, M.B., 1988. Transforming growth factor-beta is a potent

immunosuppressive agent that inhibits IL-1 dependent lymphocyte

proliferation. J. Immunol. 140, 3026–3031.

Wahl, S.M., 1992. Transforming growth factor beta (TGF-h) in inflamma-

tion: a cause and a cure. J. Clin. Immunol. 12, 61–74.

Wahl, S.M., 1994. Transforming growth factor beta: the good, the bad, and

the ugly. J. Exp. Med. 180, 1587–1590.

Yamazaki, M., Minota, S., Sakurai, H., Miyazono, K., Yamada, A.,

Kanazawa, I., Kawai, M., 1994. Expression of transforming growth

factor-beta1 and its relation to endomysial fibrosis in progressive

muscular dystrophy. Am. J. Pathol. 144, 221–226.

Ziyadeh, F.N., Hoffman, B.B., Han, D.C., Iglesias-De la Cruz, M.M., Hong,

S.W., Isono, M., Chen, S., McGowan, T.A., Sharma, K., 2000. Long-

term prevention of renal insufficiency, excess matrix gene expression,

and glomerular mesangial matrix expansion by treatment with mono-

clonal antitransforming growth factor-beta antibody in db/db diabetic

mice. Proc. Natl. Acad. Sci. U. S. A. 97, 8015–8020.