8
Lack of Skin Fibrosis in Tight Skin (TSK) Mice with Targeted Mutation in the Interleukin-4Ra and Transforming Growth Factor-b Genes Tracy McGaha, 1 Shinichiro Saito, 1 Robert G. Phelps,* Ronald Gordon,* Nancy Noben-Trauth,² William E. Paul,² and Constantin Bona Departments of Microbiology and *Pathology, Mount Sinai School of Medicine, New York, New York, U.S.A.; ²Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland, U.S.A. Scleroderma is a disorder characterized by fibrosis of the skin and internal organs and autoimmunity. Whereas the cause is unknown, interleukin-4 and transforming growth factor-b have been postulated to play a major part in the fibrosis. To investigate the part played by these cytokines, we prepared TSK/+ mice with a targeted mutation in the inter- leukin-4Ra or transforming growth factor-b genes. The breeding failed to produce TSK/+ transforming growth factor-b –/– mice so analysis of the role of transforming growth factor-b was limited to TSK/+ transforming growth factor-b +/– mice. We observed that TSK/+ interleukin-4Ra –/– did not develop der- mal thickening, and deletion of one allele of the transforming growth factor-b gene resulted in diminished dermal thickness compared with TSK/+ mice; however, the deletion of interleukin-4Ra or transforming growth factor-b had no effect on lung emphysema, which is another characteristic of TSK syndrome. Electron microscopic analysis of skin showed that the collagen fibrils in TSK/+ interleu- kin-4Ra –/– mice exhibit normal periodicity but have a smaller diameter than the fibers found in C57BL/6 mice. Analysis of skin and serum samples showed that the deletion of interleukin-4Ra or one allele of transforming growth factor-b prevented the increase of skin thickness paralleled with a decrease in the dermal hydroxyproline content and develop- ment of autoantibodies associated with TSK syn- drome. These results demonstrate the importance of interleukin-4 and transforming growth factor-b for the development of cutaneous fibrosis in vivo and suggest an important part for these cytokines in wound healing and connective tissue maintenance in general. Key words: emphysema, interleukin-4R, trans- forming growth factor-b sclerosis, tight skin. J Invest Dermatol 116:136–143, 2001 T he TSK mouse bears a chromosome 2 mutation that results in increased synthesis and accumulation of collagen in the skin and internal organs and causes lung emphysema (Green et al, 1976). It has been proposed that a partially duplicated fibrillin 1 (pdFbn- 1) gene is the mutation responsible for the scleroderma-like syndrome of TSK/+ mice (Siracusa et al, 1996). This hypothesis is strongly supported by findings demonstrating that the TSK phenotype and pdFbn-1 cosegregate (Kasturi et al, 1997); however, the mechanisms through which this mutation influences the increased synthesis and deposition of collagen that represents the endpoint of cutaneous hyperplasia is not known. The immune system, and particularly T cells, may play a part in this process as TSK/+ J H D –/– mice develop cutaneous hyperplasia and C57BL/6 pa/pa mice infused with major histocompatibility complex haplotype matched TSK/+ bone marrow cells develop a scleroderma-like syndrome (Phelps et al, 1993; Wallace et al, 1994). A possible means through which T cells may participate in the induction of fibrosis is suggested by the finding that interleukin (IL) –4 and transforming growth factor (TGF) -b, both secreted by T cells, increase the synthesis of collagen by fibroblasts (Postlethwaite, 1995). Although TSK/TSK homozygous embryos die in utero by day 9 of pregnancy, cutaneous hyperplasia in TSK/+ mice is evident 2 wk after birth. To investigate the effect of these cytokines on the development of the TSK syndrome, we prepared TSK/ + IL-4Ra –/– and TSK/+ TGF-b +/– mice. Here we show that targeted mutations in the IL-4Ra and in one TGF-b allele prevent the occurrence of cutaneous hyperplasia, and lowers the levels of dermal hydroxyproline and serum concentrations of autoantibo- dies. Interestingly, these targeted mutations do not affect the development of lung emphysema suggesting that other genes contribute to this pathologic syndrome. MATERIALS AND METHODS Mice C57BL/6 pa/pa, and TSK/+ mice were purchased from the Jackson Laboratory (Bar Harbor, ME). We used TSK/+ mice obtained by the breeding of TSK/+ 3 C57BL/6 pa/pa mice (H-2 b ) as previously described (Green et al, 1976). BALB/C IL-4Ra –/– mice (H-2 d ) mice bearing targeted mutations in IL-4Ra (Noben-Trauth et al, 1997) and C57BL/6 (H-2 b ) mice bearing a targeted mutation in TGF-b (Kulkarni et al, 1992) genes were produced at NIH Animal Facilities whereas Mov/+ mice were produced by R. Jaenisch (MIT, Boston MA) (Schnieke et al, 1983). TSK/+ IL-4Ra –/– mice were obtained by mating TSK/+ IL- Manuscript received March 28, 2000; revised October 5, 2000; accepted for publication October 10, 2000. Reprint requests to: Dr. Constantin Bona, Department of Microbiology, Box 1124, Mt. Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, U.S.A. Email: [email protected] 1 These two authors contributed equally to this work. 0022-202X/01/$15.00 · Copyright # 2001 by The Society for Investigative Dermatology, Inc. 136

Lack of Skin Fibrosis in Tight Skin (TSK) Mice with Targeted Mutation in the Interleukin-4Ralpha and Transforming Growth Factor-beta Genes

Embed Size (px)

Citation preview

Lack of Skin Fibrosis in Tight Skin (TSK) Mice with TargetedMutation in the Interleukin-4Ra and Transforming GrowthFactor-b Genes

Tracy McGaha,1 Shinichiro Saito,1 Robert G. Phelps,* Ronald Gordon,* Nancy Noben-Trauth,²William E. Paul,² and Constantin BonaDepartments of Microbiology and *Pathology, Mount Sinai School of Medicine, New York, New York, U.S.A.; ²Laboratory of Immunology,

National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Maryland, U.S.A.

Scleroderma is a disorder characterized by ®brosis ofthe skin and internal organs and autoimmunity.Whereas the cause is unknown, interleukin-4 andtransforming growth factor-b have been postulatedto play a major part in the ®brosis. To investigatethe part played by these cytokines, we preparedTSK/+ mice with a targeted mutation in the inter-leukin-4Ra or transforming growth factor-b genes.The breeding failed to produce TSK/+ transforminggrowth factor-b ±/± mice so analysis of the role oftransforming growth factor-b was limited to TSK/+transforming growth factor-b +/± mice. We observedthat TSK/+ interleukin-4Ra ±/± did not develop der-mal thickening, and deletion of one allele of thetransforming growth factor-b gene resulted indiminished dermal thickness compared with TSK/+mice; however, the deletion of interleukin-4Ra ortransforming growth factor-b had no effect on lungemphysema, which is another characteristic of TSK

syndrome. Electron microscopic analysis of skinshowed that the collagen ®brils in TSK/+ interleu-kin-4Ra ±/± mice exhibit normal periodicity buthave a smaller diameter than the ®bers found inC57BL/6 mice. Analysis of skin and serum samplesshowed that the deletion of interleukin-4Ra or oneallele of transforming growth factor-b prevented theincrease of skin thickness paralleled with a decreasein the dermal hydroxyproline content and develop-ment of autoantibodies associated with TSK syn-drome. These results demonstrate the importance ofinterleukin-4 and transforming growth factor-b forthe development of cutaneous ®brosis in vivo andsuggest an important part for these cytokines inwound healing and connective tissue maintenance ingeneral. Key words: emphysema, interleukin-4R, trans-forming growth factor-b sclerosis, tight skin. J InvestDermatol 116:136±143, 2001

The TSK mouse bears a chromosome 2 mutation thatresults in increased synthesis and accumulation ofcollagen in the skin and internal organs and causeslung emphysema (Green et al, 1976). It has beenproposed that a partially duplicated ®brillin 1 (pdFbn-

1) gene is the mutation responsible for the scleroderma-likesyndrome of TSK/+ mice (Siracusa et al, 1996). This hypothesis isstrongly supported by ®ndings demonstrating that the TSKphenotype and pdFbn-1 cosegregate (Kasturi et al, 1997); however,the mechanisms through which this mutation in¯uences theincreased synthesis and deposition of collagen that represents theendpoint of cutaneous hyperplasia is not known.

The immune system, and particularly T cells, may play a part inthis process as TSK/+ JHD ±/± mice develop cutaneous hyperplasiaand C57BL/6 pa/pa mice infused with major histocompatibilitycomplex haplotype matched TSK/+ bone marrow cells develop ascleroderma-like syndrome (Phelps et al, 1993; Wallace et al, 1994).A possible means through which T cells may participate in the

induction of ®brosis is suggested by the ®nding that interleukin (IL)±4 and transforming growth factor (TGF) -b, both secreted by Tcells, increase the synthesis of collagen by ®broblasts (Postlethwaite,1995). Although TSK/TSK homozygous embryos die in utero byday 9 of pregnancy, cutaneous hyperplasia in TSK/+ mice isevident 2 wk after birth. To investigate the effect of these cytokineson the development of the TSK syndrome, we prepared TSK/+ IL-4Ra ±/± and TSK/+ TGF-b +/± mice. Here we show thattargeted mutations in the IL-4Ra and in one TGF-b allele preventthe occurrence of cutaneous hyperplasia, and lowers the levels ofdermal hydroxyproline and serum concentrations of autoantibo-dies. Interestingly, these targeted mutations do not affect thedevelopment of lung emphysema suggesting that other genescontribute to this pathologic syndrome.

MATERIALS AND METHODS

Mice C57BL/6 pa/pa, and TSK/+ mice were purchased from theJackson Laboratory (Bar Harbor, ME). We used TSK/+ mice obtained bythe breeding of TSK/+ 3 C57BL/6 pa/pa mice (H-2b) as previouslydescribed (Green et al, 1976). BALB/C IL-4Ra ±/± mice (H-2d) micebearing targeted mutations in IL-4Ra (Noben-Trauth et al, 1997) andC57BL/6 (H-2b) mice bearing a targeted mutation in TGF-b (Kulkarniet al, 1992) genes were produced at NIH Animal Facilities whereas Mov/+mice were produced by R. Jaenisch (MIT, Boston MA) (Schnieke et al,1983). TSK/+ IL-4Ra ±/± mice were obtained by mating TSK/+ IL-

Manuscript received March 28, 2000; revised October 5, 2000; acceptedfor publication October 10, 2000.

Reprint requests to: Dr. Constantin Bona, Department of Microbiology,Box 1124, Mt. Sinai School of Medicine, One Gustave L. Levy Place, NewYork, NY 10029, U.S.A. Email: [email protected]

1These two authors contributed equally to this work.

0022-202X/01/$15.00 ´ Copyright # 2001 by The Society for Investigative Dermatology, Inc.

136

4Ra +/± mice and TSK/+ TGF-b +/± mice were obtained by breedingTSK/+ and TGF-b +/± mice. Fibroblast lines were obtained from 9 d oldC57BL/6 pa/pa, TSK/+, and Mov/Mov embryos as previously described(Kasturi et al, 1997).

Reagents The Fbn-1 fusion protein was prepared as previously described(Murai et al, 1998). Other reagents were purchased from Sigma (St Louis,MO) unless otherwise noted.

Identi®cation of TSK mutant and of TGF-b and IL-4R nullalleles Genomic DNA were prepared from tail biopsies with QuiagenTissue Kits (Quiagen, Hilden, Germany). Polymerase chain reactionanalysis was preformed for identi®cation of pdFbn-1 gene with a forwardprimer of 5¢-GTTGGCAACTATACCTGCAT-3¢ and a reverse primer of5¢-CCTTTCCTGGTAACATAGGA-3¢. Cycling conditions were 95°Cfor 1 min primary denaturation, then 95°C, 59°C for 1 min, 72°C for 45 sfor 30 cycles in a 50 ml volume. Polymerase chain reaction for the wild-typeTGF-b allele was carried out with the forward primer 5¢-TCACCCGCG-TGCTAATGGTGGACCGC-3¢ and reverse primer of 5¢-ACACCTTC-CATTCTCTTGAGCTGGG-3¢, and for the disrupted TGF-b allele withthe forward primer of 5¢-CATGGAGCTGGTGAAACGGAAGCGC-3¢and the reverse primer of 5¢-TCCATCTGCACGAGACTAGT-3¢ underthe cycle conditions described above.

Polymerase chain reaction for the wild-type IL-4R allele was done withthe forward primer of 5¢-AATGTGACCTACAAGGAACC-3¢ andreverse primer of 5¢-GGACTCCACTCACTCCAG-3¢, and for thedisrupted IL-4R allele with the forward primer of 5¢-CTTGGGTGGAG-AGGCTATTC-3¢ and the reverse primer of 5¢-AGGTGAGATGACAG-GAGATC-3¢ under the same cycle conditions.

Southern blotting Genomic DNA were prepared from ®broblastsobtained from 9 d old embryos produced by the breeding of Mov/+ mice.Samples (10 mg) of genomic DNA were digested with the appropriaterestriction enzymes, separated by electrophoresis on a 1% agarose gel, andblotted on to nitrocellulose ®lters. The ®lters were hybridized with a nicktranslated 32P-labeled a1 collagen type 1 probe at 42°C in hybridizationsolution containing 50% formamide and ®nally washed in 0.2 3 sodiumcitrate/chloride buffer containing 0.1% sodium dodecyl sulfate at 65°C.

Light and electron microscopy Skin samples (4.0 cm2) were removedfrom the dorsal side immediately below the neck in a manner thatminimized stress. The samples were ®xed for 16 h in buffered salinecontaining formalin, cut into 2±3 mm wide longitudinal strips, dehydrated,and embedded in paraf®n and stained with hematoxylin±eosin according toroutine histologic methods. The thickness of the skin was determined bymeasuring it from the top of the granular layer to the junction between thedermis and subcutaneous fat on hematoxylin±eosin-stained sections.

Skin tissues for electron microscopy were processed following standardtechniques. Each sample was assigned a random number and the diameterof the collagen ®bers was measured blindly as previously described (Phelpset al, 1993).

In¯ated lungs after harvesting were ®xed and processed by the samemethods described above. The extent of airspace was analyzed onhematoxylin±eosin-stained sections.

Determination of skin hydroxyproline content Punch biopsies(6 mm diameter) from shaved skin samples were treated withchloroform/methanol (2:1 vol/vol) to remove fat and dried by Speedvaccentrifugation. Dried samples were weighed and acid hydrolyzed for 24 h at110°C. The hydroxy lysates were dried, redissolved in 200 ml of water and®ltered through millipore ®lters. Sample aliquots of 20 ml were diluted 10-fold and used for the analysis of amino acid composition in a Hewlett-Packard Amino Acid Analyzer. The quantity of hydroxyproline isexpressed as (mol per mg dry skin (Hatakeyama et al, 1996).

Quantitation of serum anti-topoisomerase I and anti-®brillin 1antibodies Serum anti-topoisomerase I and anti-®brillin 1 levels weremeasured by radioimmunoassay as previously described (Muryoi et al, 1992;Murai et al, 1998). Brie¯y, microtiter plates were coated with 3 mg per ml ofpuri®ed topoisomerase I (GibcoBRL, Gaithersburg, MD) or 1 mg per ml ofFbn-1 fusion protein in carbonate buffer, pH 9.6, washed with phosphate-buffered saline, blocked with 1% bovine serum albumin in phosphate-buffered saline and incubated for 90 min at room temperature with a 1:30dilution of serum samples. After extensive washing, the plates wereincubated with 50,000 cpm of 125I-labeled rat anti-mouse k per well for90 min. Bound radioactivity was measured in a g spectrometer.

Cell culture and measurement of collagen synthesis C57BL/6 pa/pa and TSK/+ primary ®broblast lines were maintained in DMEMsupplemented with 100 U per ml penicillin 100 mg per ml streptomycin and10% fetal bovine serum (Mediatech, Herndon, VA). Collagen productionwas measured using a modi®ed version of the protocol described byPeterkofsky and Diegelmann (1970). Brie¯y, ®broblasts were grown tocon¯uence in 75 cm2 ¯asks, detached via trypsinization and plated at adensity of 105 cells per well in 24 well plates. The cells were washed withphosphate-buffered saline and the media was replaced with proline freeDMEM supplemented with 3 mCi per well 3H-proline (Amersham,Piscataway, NJ), 50 mg per ml ascorbic acid, and b-aminopropionitrileand with 400 U per ml recombinant murine IL-4 (Pharmingen, San Diego,CA), 10 ng per ml recombinant human TGF-b1 or 0.5±50 ng per ml IL-13(R&D Systems, Minneapolis, MN). After a 24 h incubation, the mediawere collected for determination of collagen synthesis and the cell numberswere counted. To measure collagen synthesis, 100 ml aliquots of mediafrom labeled ®broblast were incubated at 37°C for 18 h in either thepresence or absence of highly puri®ed bacterial collagenase (Sigma) at aconcentration of 200 U per aliquot. The samples were then extensivelydialyzed in microdialysis chambers (Pierce, Rockford, IL) againstphosphate-buffered saline at 4°C to remove unincorporated 3H-prolineand digested collagen. The total counts per sample were then determinedutilizing a liquid scintillation counter. Biosynthetic labeling of collagen wasestimated by subtracting the cpm of the collagenase-digested dialysisretentate from the cpm of the dialysis retentate which had no collagenaseadded.

Statistical analysis Statistical signi®cance between groups was measuredusing the unpaired Student's t test. p < 0.05 was considered to besigni®cant.

RESULTS

Increase of collagen synthesis by ®broblast incubated withIL-4 and TGF-b IL-4 and TGF-b have been reported toupregulate the expression of collagen genes (Postlethwaite, 1995).We studied the synthesis of collagen by ®broblast lines fromC57BL/6 pa/pa and TSK/+ mice cultured for 20 h in media aloneor in the presence of 400 U per ml of IL-4 or 10 ng per ml of TGF-b (Fig 1A). Collagen synthesis by normal ®broblasts was increased2-fold upon the addition of IL-4 or TGF-b. In addition, there wasa highly signi®cant increase of collagen synthesis by TSK/+®broblast after incubation with either cytokine; however, nosigni®cant increase of collagen synthesis was observed afterincubation of ®broblasts with 0.5±50 ng per ml of IL-13 (data notshown). This result is somewhat unexpected as IL-4 and IL-13utilize the IL-4Ra chain and have generally been shown to possesssimilar functions in vivo.

The speci®city of the assay was determined by studying thesynthesis of collagen by Mov/Mov ®broblasts. The genotypes of

Figure 1. Synthesis of collagen by normal and TSK/+ ®broblastsupon incubation with IL-4 and TGF-b. Bars represent the mean valuefor quadruplicate samples of 105 ®broblast +/± the SD. The experimentwas repeated three times with similar results.

VOL. 116, NO. 1 JANUARY 2001 AUTOIMMUNITY AND CYTOKINES IN SKIN SCLEROSIS 137

these ®broblasts were determined by southern blotting (data notshown). As can be seen in Fig 1, Mov/Mov ®broblasts failed tosynthesize collagen consistent with the ®nding that a retroviralinsertion has occurred in the a1 collagen type I gene inactivating it(Schnieke et al, 1983). These results show that both IL-4 and TGF-b upregulate the expression of collagen genes by ®broblasts fromboth TSK/+ and normal mice. In addition, these resultsdemonstrate that TSK/+ ®broblasts secrete signi®cantly higherlevels of collagen when compared with C57BL/6 ®broblasts in thebasal state.

Crossing of TSK/+ mice with mice bearing targetedmutations in the IL-4R and TGF-b genes Heterozygous

viable TSK/+ pa/pa mice were obtained by crossing TSK/+ pa/pamice with C57BL/6 pa/pa mice. The TSK mutation was originallydescribed in a B10D2 (H-2d) mouse, which was maintained inC3HDi (H-2b) background and subsequently transferred to theC57BL/6 pa/pa (H-2b) background (Green et al, 1976).Occurrence of cutaneous hyperplasia in these different geneticbackgrounds, as well as in TSK/+ vit/vit (Hatakeyama et al, 1996)or TSK/+ NZB/+ mice (Bocchieri et al, 1993) demonstrates thatthe occurrence of TSK syndrome is independent of geneticbackground.

TSK/+ mice bearing a targeted mutation in the TGF-b genewere obtained by crossing TSK/+ pa/pa mice with TGF-b +/±mice on the C57BL/6 (H-2b) background, and TSK/+ IL-4R ±/±

Figure 2. Histopathology of the skin from TSK/+ IL-4R+/±, TSK/+ IL-4R±/±, or TSK/+ TGF-b +/± mice. Paraf®n-embedded sections werestained with hematoxylin±eosin. Scale bar: 100 mM.

Figure 3. Histopathology of the lung from TSK/+ IL-4R+/±, TSK/+ IL-4R ±/±, and TSK/+ TGF-b +/± mice. Paraf®n-embedded sectionswere stained with hematoxylin±eosin. Scale bar: 1.6 mM. Lung sections from two TSK/+ TGF-b +/± mice are presented. As can be seen in one mouse,strong cellular in®ltration has occurred in the interstitium (bottom right).

138 MCGAHA ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

mice were obtained from sister±brother crossing of TSK/+ IL-4Ra +/± (H-2db) mice. The progeny from both crossings weregenotyped by polymerase chain reaction as described in Materialsand Methods.

From the breeding of TSK/+ IL-4R +/± mice we studied 71mice: 36 expressing the TSK mutation (TSK/+) and 35 lacking themutation (+/+). Among the 36 TSK/+ mice, nine were IL-4Ra ±/±, 23 were IL-4Ra +/±, and four were IL-4Ra +/+.

From the breeding of TSK/+ TGF-b +/± mice we studied 85progeny: 43 expressed the TSK mutation (TSK/+) and 42 did not(+/+). Among the 43 mice bearing the TSK mutation, 37 wereTGF-b +/±, six were TGF-b +/+, and none exhibited the TGF-b ±/± genotype. It is noteworthy that among 42 mice lacking theTSK mutation (±/±) we obtained only three TGF-b ±/± mice,which is in agreement with previous ®ndings that indicate a highmortality rate (> 50%) in utero for mice lacking a functional TGF-bgene (Christ et al, 1994). The absence of the TSK/+ TGF-b ±/±genotype in our crossings may suggest that unknown epistatic geneinteractions prevent the development of mice possessing thisgenotype.

Histopathology of skin and lung of TSK/+ mice expressinga targeted mutation in the IL-4R or TGF-bgene Histopathology of the skin and lung was studied in 3 moold mice. Figure 2 shows skin alterations in TSK/+ mice and thenormal structure of skin of C57BL/6 pa/pa mice. TSK/+ miceexhibit cutaneous hyperplasia characterized by increased depositionof collagenous material in the super®cial and deep dermis extendingaround adnexa and the paniculum carnosum associated with thedisappearance of fat tissue and a reduced number of hair follicles. Incontrast, TSK/+ IL-4Ra ±/± mice show lack of dermal thickening,whereas TSK/+ IL-4Ra +/± mice showed a moderate thickeningof the skin in which the fatty tissue is preserved (Fig 3). In the same®gure the skin sections of the two TSK/+ TGF-b +/± micepresented show that skin thickness is slightly decreased compared

with TSK/+ mice with preservation of fat tissue and hair follicles;however, it should be pointed out that in one mouse cellularin®ltration in the deep dermis was observed (Fig 2, center bottompicture). Only one TSK/+ TGF-b +/+ mouse was analyzed forTSK syndrome and showed a skin thickness of 200 mm, ahydroxyproline concentration of 1054 mmol per mg,autoantibody levels of 4448 and 9851 cpm for anti-topoisomeraseI and anti-®brillin 1, respectively, and the presence of pulmonaryemphysema.

The lungs of C57BL/6 mice appeared normal, with small alveoliof approximately the same size. By contrast, TSK/+ lungs displayedenlarged alveoli that were three to four times larger than normalmice (Fig 3) in agreement with the description of Rossi et al(1984). TSK/+ IL-4Ra ±/± and TSK/+ TGF-b +/± mice, likeTSK/+ and TSK/+ IL-4Ra +/± mice showed a histologic pictureof emphysema (Fig 3).

Electron microscopy analysis Electron microscopicexamination of the skin con®rmed the observations by lightmicroscopy and allowed a more detailed analysis. In contrast toC57BL/6 pa/pa mice (Fig 4A), where there was no excessivedeposition of collagen in the deep dermis and regular collagenbundles were found, the skin from TSK/+ mice (Fig 4B) showedabundant irregular and wavy collagen bundles. TSK/+ IL-4Ra ±/±and TSK/+ TGF-b +/± skin did not exhibit any alteration of thecollagen bundles. Figure 5 shows that in C57BL/6 pa/pa mice,large ®bers (70±130 nm) were predominant whereas in TSK/+skin, small diameter ®bers (50±100 nm ®bers) were predominant.In studies carried out on skin biopsies of human sclerodermapatients the predominance of small diameter ®bers was consideredpathognomonic for neo®brogenesis (Flieischmajer et al, 1991).Predominance of small diameter ®bers was observed in TSK/+ IL-4Ra ±/± skin (70±110 nm) and TSK/+ IL-4Ra +/± skin (70±100 nm). TSK/+ TGF-b +/± skin also displayed small diameter®bers (40±90 nm). Thus, electron microscopic analysis and size of

Figure 4. Electron microscopy of dermis and collagen bundles in the skin of C57BL/6 pa/pa (A), TSK/+ (B), TSK/+ IL-4R ±/± (C), TSK/+ IL-4R +/±(D), and TSK/+ TGF-b +/± (E) mice.

VOL. 116, NO. 1 JANUARY 2001 AUTOIMMUNITY AND CYTOKINES IN SKIN SCLEROSIS 139

collagen ®bers revealed that the IL-4Ra ±/± and TGF-b +/±genotype did not rescue all the skin abnormalities of TSK/+ mice.

TSK syndrome in TSK/+ mice bearing targeted mutationsin IL-4R or TGF-b genes Hatakeyama et al (1996) have shownthat there is a striking correlation between the histologic andbiochemical alterations in the skin of TSK/+ mice and theoccurrence of anti-topoisomerase I antibodies. We usedHatakeyama's parameters to de®ne the TSK syndromedetermining the skin thickness, concentration of dermalhydroxyproline, and the presence of anti-topoisomerase Iautoantibodies. Murai et al (1998) showed that anti-®brillin 1autoantibodies are present in TSK/+ mice, but not in C57BL/6pa/pa mice; thus, anti-®brillin 1 autoantibodies were also analyzedin our study. Table I shows that in TSK/+ mice there is asigni®cant increase in skin thickness (p = 0.02), hydroxyprolinecontent (p = 0.01), anti-topoisomerase I antibodies (p = 0.002), andanti-®brillin 1 antibodies (p = 0.004) compared with C57BL/6 pa/pa mice. In TSK/+ IL-4Ra ±/± mice, the thickness of the skin and

dermal hydroxyproline content were similar to C57BL/6 pa/pamice and were considerably reduced when compared with TSK/+mice. In contrast, in TSK/+ IL-4Ra +/± mice the skin thicknesswas increased compared with normal mice (p = 0.018); there wasalso an increase in hydroxyproline content, although it was notstatistically signi®cant (p = 0.26); however, both skin thickness andhydroxyproline content of TSK/+ IL-4Ra +/± skin wassigni®cantly greater than that of TSK/+ IL-4Ra ±/± skin. Withrespect to autoantibodies, TSK/+ IL-4Ra +/± mice show asigni®cant increase of anti-topoisomerase I compared withnormal mice and TSK/+ IL-4Ra ±/± mice; however, TSK/+ IL-4Ra +/± mice did not display a signi®cant increase in anti-®brillin1 antibodies compared with either normal mice or TSK/+ IL-4Ra ±/± mice.

As the comparison of TSK syndrome parameters between TSK/+ IL-4Ra ±/± mice and C57BL/6 mice is biased because ofgenetically different backgrounds we compared the TSK syndromebetween TSK/+ IL-4Ra ±/± and TSK/+ IL-4Ra +/+ mice. Wefound a signi®cant decrease in the skin thickness (p = 0.02), dermal

Figure 5. Histogram showing the distribution ofdiameter of collagen ®bers present in the mid-der-mis of C57BL/6 pa/pa (A), TSK/+ (B), TSK/+ IL-4R ±/± (C), TSK/+ IL-4R +/± (D), andTSK/+ TGF-b +/± (E) mice.

Table I. TSK syndrome in TSK/+IL-4Ra±/± and TSK/+TGF-b+/± micea

Autoantibodies speci®c for

Mouse strain N Skin thickness(mm)

Hydroxyproline content(mmol/mg dry skin)

Topo-I(cpm)

Fbn-I(cpm)

C57BL/6 pa/pa 6 132 6 42 xp xxp 518 6 88 xp xxp 512 6 472 xp xxp 1379 6 12 xp xxp

TSK/+ pa/+ 13 228 6 72 0.02 1035 6 310 0.01 2733 6 337 0.002 11196 6 49 0.004

TSK/+IL-4Ra+/± 9 243 6 88 0.018 } 0.02804 6 167 0.26 } 0.005

1180 6 649 0.07 } 0.063409 6 25 0.08 } 0.8

TSK/+IL-4Ra±/± 9 135 6 31 0.2 492 6 244 0.12 607 6 421 0.66 366 6 18 0.03

+/+TGF-b+/± 3 120 6 18 0.6 } 0.019441 6 100 0.4 } 0.4

737 6 30 0.37 } 0.854967 6 10 0.026 } 0.14

TSK/+TGF-b+/± 16 164 6 78 0.16 442 6 217 0.16 446 6 248 0.77 2850 6 14 0.06

aValues represent mean values 6 standard deviation (SD). xp, value comparing various strains to wild type C57BL/6 pa/pa mice; xxp, value comparing genotypicallysimilar strains.

140 MCGAHA ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

hydroxyproline content (p = 0.05), and anti-topoisomerase Iantibodies (p = 0.06), but not in the case of anti-®brillin 1antibodies (p = 0.8) in TSK/+ IL-4Ra ±/± compared with TSK/+ IL-4Ra +/± mice. In addition, both TSK/+ IL-4Ra ±/± andTSK/+ IL-4Ra +/± mice develop emphysema.

TSK/+ TGF-b +/± mice displayed no signi®cant increase in skinthickness (p = 0.144), dermal hydroxyproline content (p = 0.16),anti-topoisomerase I antibodies (p = 0.07), or anti-®brillin 1antibodies (p = 0.6) compared with normal mice; however, wefound signi®cant differences between the skin thickness whencomparing TSK/+ TGF-b +/± with +/+ TGF-b +/± mice(p = 0.019).

Thus in the absence of IL-4Aa and when only one copy of afunctional TGF-b gene is present several of the important aspects ofthe TSK syndrome (skin thickness, increased hydroxyproline, andincreased anti-topoisomerase I antibodies) were decreased.

DISCUSSION

In this study we examined the part that TGF-b and IL-4 play in thedevelopment of TSK syndrome. Initially, we demonstrated thatincreased synthesis of collagen by normal and TSK/+ ®broblastsoccurred upon short exposure to IL-4 and TGF-b. This outcome isin agreement with several reports that have found both IL-4 andTGF-b to upregulate expression of the type 1 collagen genes(Fertin et al, 1991; Serpier et al, 1997; Cotton et al, 1998; Ichiki et al,1997; Inagaki et al, 1994). Similarly, we demonstrated that TSK/+®broblasts spontaneously produce more collagen than C57BL/6pa/pa derived ®broblasts and a greater increase in collagenproduction is observed in response to stimulation with either IL-4 or TGF-b. Serpier et al (1997) made similar observations whenthey found that ®broblasts from scleroderma patients produce morecollagen than their normal counterparts, express higher levels of IL-4Ra, and produced greater levels of collagenase-digestible proteinafter stimulation with IL-4. In addition, scleroderma ®broblastshave been reported to express higher levels of TGF-bRI and TGF-bRII and to bind higher levels of 125I-labeled TGF-b than normal®broblasts (Kawakami et al, 1998) suggesting that scleroderma®broblasts are more sensitive to pro®brotic stimuli from TGF-b orIL-4.

There are no available data on the in vivo effect of these cytokineson the ®brotic processes. To address this question, we generatedTSK/+ IL-4Ra ±/± and TSK/+ TGF-b +/± mice. It is note-worthy that whereas TSK/+ mice exhibiting a targeted mutation inTGF-b genes were generated in the C57BL/6 (H-2b) background,TSK/+ IL-4Ra ±/± mice had a mixed genetic background (H-2d,H-2b, and H-2db) as illustrated by coat color: two white, threeblack, and six agouti. We believe, however, that the mixed geneticbackground did not alter the development of TSK syndrome forthree reasons: (i) It was shown that the occurrence of cutaneoushyperplasia is solely due to the TSK mutation and is not affected byvarious genetic backgrounds (Green et al, 1976); (ii) it was shownthat TSK/+ JHD ±/± (Kasturi et al, 1997) and TSK/+ RAG2 ±/±mice (Siracusa et al, 1998), which were bred in a manner similar tothat described here and have the same genetic background as theIL-4Ra ±/± mice, developed cutaneous hyperplasia; and (iii) asTSK/+ IL-4Ra ±/± mice were not obtained on the C57BL/6background, the results in our study were always compared withTSK/+ IL-4Ra +/± mice, which have a similar genetic back-ground.

We showed that disruption of the IL-4Ra gene results inreduced ®brotic development in TSK/+ mice. The lack ofcutaneous hyperplasia in TSK/+ IL-4Ra ±/± mice was observedthroughout their life (thickness of skin was 151 6 51 mm in 3 moold mice and 150 6 70 mm in 6 mo old mice) indicating that IL-4Ra signaling is important in the development of ®brosis in TSK/+ mice. In the case of TSK/+ TGF-b +/± mice, we also observed adecrease in skin thickness. It was originally reported that TGF-b +/± mice do not exhibit an altered phenotype (Shull et al, 1992;Christ et al, 1994); however, recent studies show a defect in wound

healing in TGF-b +/± mice (Ashcroft et al, 1999) indicating aphenotypically dominant effect in heterozygous TGF-b KO mice.This may have particular relevance to our observations of decreasedskin thickness in TSK/+ TGF-b +/± mice as it was previouslyshown that TGF-b transcripts were signi®cantly elevated in theskin of TSK/+ mice when compared with pa/pa mice (Phelps et al,1993). It is possible that the development of cutaneous hyperplasiain TSK/+ requires a certain threshold level of TGF-b, which is notachieved by the disruption of one of the TGF-b alleles. This wouldbe consistent with the observation that TGF-b transcripts inheterozygous animals was approximately half of the level of TGF-b+/+ mice (Kulkarni et al, 1993).

A morphometric study of collagen ®bers shows that in the skin ofTSK/+ mice, TSK/+ IL-4Ra +/±, and TSK/+ TGF-b +/± micesmall diameter ®bers predominate. This outcome suggests that thelack of TGF-b or IL-4Ra does not totally correct the defectsassociated with TSK syndrome and is supported by earlier studieswhere small diameter ®bers of collagen in the skin were interpretedas a sign of neo®brogenesis (Flieischmajer et al, 1991). These resultswere in sharp contrast to the diameter of ®brils in C57BL/6 pa/pacontrol mice in which the large diameter ®bers were dominant(Fig 5).

Because of these results, we examined the effect of disruption ofIL-4Ra and TGF-b genes on the TSK syndrome. Compared withTSK/+ mice, TSK/+ IL-4Ra ±/± and TSK/+ TGF-b +/± micedid not develop autoantibodies against the scleroderma antigenstopoisomerase I (Shero et al, 1986) and ®brillin 1 (Tan et al, 1999).In the case of TSK/+ IL-4Ra ±/± mice, this may be explained asIL-4 signaling plays an important part in the development of TH

cells cooperating with B cells in the production of autoantibodies.In the case of TGF-b de®cient mice, however, the lack of thiscytokine may skew the immune responses towards a Th1 typeresponse and away from antibody production. Li et al (1999) foundthat the neutralization of TGF-b aided mice in the clearance of thepathogen Leishmania major via enhanced cell-mediated immunity.In addition, it has been shown that TGF-b decreases interferon-gand tumor necrosis factor-a production by T cells (Holter et al,1994; Erard et al, 1999), and the deletion of TGF-b inducesin¯ammatory reactions (Shull et al, 1992). Therefore, it is likely thatthe animals with a targeted disruption in the TGF-b gene produce amore in¯ammatory cytokine pro®le and, thus, have a lowerantibody titer.

It was very striking that the disruption of these two genes doesnot affect the development of lung emphysema. This is interestingbecause ®brillin 1 is an important component of micro®bers in thelung (Sakai et al, 1986). In principle, the mutated ®brillin 1 in TSK/+ mice is likely to be responsible for the alveolar wall abnormalities;however, the presence of emphysema with a lack of skin ®brosisindicates a disassociation of the effect of disruption of IL-4Ra andTGF-b genes on cutaneous hyperplasia and emphysema. This resultstrongly suggests that factors other than IL-4 and TGF-b areresponsible for the emphysema. This concept is supported by ourown previous results demonstrating that Tg mice expressing thepartially duplicated Fbn-1 gene also develop a scleroderma-likesyndrome and, interestingly, newborn mice injected with a plasmidencoding the mutated Fbn-1 gene develop localized sclerodermabut lack pulmonary emphysema (Saito et al, 2000).

These results suggest that IL-4 and TGF-b in¯uence ®brosis invivo. Nevertheless it is unclear what the source of these cytokinesmay be as TSK/+ mice begin to exhibit skin ®brosis by 2 wk ofage. There is little information on the synthesis of IL-4 and TGF-bin embryonic life. Eosinophils, which are able to produce IL-4, canbe excluded because no eosinophilic in®ltration has been observedin the dermis of TSK/+ mice (Green et al, 1976); therefore, othercell types are likely to be responsible for the cutaneous hyperplasia.For instance mastocytes, which are present in increased numbers inthe skin of TSK/+ mice (Green et al, 1976; Phelps et al, 1993;Ridge et al, 1996), CD4+ T cells, and the ®broblast themselves havebeen reported to be capable of synthesizing IL-4 and TGF-b. In thecase of ®broblasts, it has been demonstrated that scleroderma

VOL. 116, NO. 1 JANUARY 2001 AUTOIMMUNITY AND CYTOKINES IN SKIN SCLEROSIS 141

®broblasts spontaneously produce high levels of IL-4 in vivo and invitro in the absence on stimuli from other cell types (White et al,1992; Salmon-Ehr et al, 1996). Whereas the transcript for TGF-bwas found to be increased in the skin of TSK/+ mice (Phelps et al,1993), there is no information whether the IL-4 transcript isincreased. The evidence for an in vivo effect of IL-4 in TSKcutaneous hyperplasia is provided by our observation that TSK®broblasts synthesize more collagen in response to IL-4 stimulationwhen compared with C57BL/6 ®broblasts and Ong et al (1998)reported that the injection of anti-IL-4 antibodies in TSK neonatesprevented cutaneous hyperplasia.

As stated above, both IL-4 and TGF-b upregulate the expressionof collagen genes. This means that in mice with targeted mutationsin IL-4Ra, the TGF-b genes should be able to compensate for theIL-4 defect. Likewise, in TGF-b +/± mice IL-4 should compensatefor the TGF-b defect; however, in TSK/+ IL-4Ra ±/± and, to alesser extent, in TSK/+ TGF-b +/± mice, we observed that atargeted mutation prevented cutaneous hyperplasia. Two possiblehypothesis can be suggested to explain these results. First, theproduction of TGF-b requires IL-4. Elovic et al (1999) have shownthat the presence of IL-4 is necessary for the production of TGF-b.Additionally, it has been shown that IL-4 upregulates theproduction of TGF-b in T cells whereas TGF-b downregulatesthe production of IL-4 (Holter et al, 1994; Seder et al, 1998; Elovicet al, 1999). Therefore, IL-4 may serve as a physiologic molecularswitch for gene expression of TGF-b. In this scenario IL-4 wouldbe the inducer of ®brosis by elevating TGF-b production;however, once TGF-b was induced it would act as a positiveregulator of its own expression via an autocrine pathway(Kawakami et al, 1998; Seder et al, 1998) while simultaneouslyinhibiting IL-4 production. Therefore, the deletion of eithercytokine would have the net effect of inhibiting ®brosis. Second, itis possible that a common molecule in IL-4 and TGF-b signalingpathways may be involved in the regulation of collagen geneexpression. This scenario suggests that the IL-4 and TGF-bsignaling intermediates act in a synergistic fashion to upregulateextracellular matrix production. Support for this hypothesis wasprovided by Nakashimia et al (1999) who found that stimulationwith leukemia inhibitory factor and bone morphogenic protein-1results in the formation of a Smad1/STAT3 signaling complex andthis interaction is necessary for the development of astrocytes fromneural progenitor cell. In this way the combination of the twocytokines may be necessary to generate the strength of signalnecessary to overcome a biologic threshold and induce the®brogenic phenotype in TSK/+ mice. Therefore, the lack ofeither cytokine would result in diminishing signal strength to apoint where it could no longer overcome this threshold andeffectively abrogate the ®brosis observed in TSK/+ mice.

In summary, our results demonstrate the in vivo effect of IL-4 andTGF-b signaling on skin sclerosis in mice as illustrated in the lack ofcutaneous hyperplasia in TSK/+ IL-4Ra ±/± and, to a lesserextent, in TSK/+ TGF-b +/± mice. These results may be exploitedfor the development of new therapeutic approaches for sclerodermaand suggest a major part for these cytokines in ®brosis and woundhealing.

This work was supported by a grant from the Scleroderma Foundation and NIAID-

NIH grant PO1AI24671-12.

REFERENCES

Ashcroft GS, Yang X, Glick AB, et al: Mice lacking Smad3 show accelerated woundhealing and impaired local in¯ammatory response. Nature Cell Biol 1:260±266,1999

Bocchieri MH, Christner PJ, Henriksen PD, Jimenez SA: Immunologicalcharacterization of (Tight Skin/NZB) F1 hybrid mouse with connectivetissue and autoimmune features resembling human systemic sclerosis. JAutoimmun 6:337±351, 1993

Christ M, McCartney-Francis NL, et al: Immune dysregulation in TGF-b1-de®cientmice. J Immunol 153:1937±1945, 1994

Cotton SA, Herrick AL, Jayson MIV, Freemont AJ: TGF-bÐa role in systemicsclerosis? J Pathol 184:4±6, 1998

Elovic AE, Ohyama H, Sauty A, et al: IL-4-dependent regulation of TGF-b andTGF-b 1 expression in human eosinophils. J Immunol 160:6121±6127, 1999

Erard F, Garcia-Sanz JA, Moriggl R, Wild M-T: Presence or absence of TGF-bdetermines IL-4-induced generation of type 1 or type 2, CD8 T cell subsets. JImmunol 162:209±214, 1999

Fertin C, Nicolas JF, Gillery P, Kalis B, Bancherau J, Maquart F-X: Interleukin-4stimulates collagen synthesis by normal and sclerodermal ®broblast in dermalequivalents. Cell Mol Biol 37(8):823±829, 1991

Flieischmajer R, Jacobs L, Schwartz E, Sakai LY: Extracellular micro®brils areincreased in localized and systemic scleroderma skin. Lab Invest 64:791±798,1991

Green MC, Sweet HO, Bunker LE: Tight-skin, a new mutation of the mousecausing excessive growth of connective tissue and skeleton. Am J Pathol82:493±507, 1976

Inagaki Y, Truter S, Ramirez F: Transforming growth factor b stimulates a2(I)collagen gene expression through a cis-acting element that contains an SP-1binding site. J Biol Chem 269:14828±14834, 1994

Hatakeyama A, Kasturi KN, Wolf I, Phelps RG, Bona CA: Correlation between theconcentration of serum anti-topoisomerase 1 autoantibodies and histologicaland biochemical alterations in the skin of tight skin mice. Cell Immunol167:135±140, 1996

Holter W, Kalthoff FS, Pickl WF, Ebner C, Majdic O, Kraft D, Knapp W:Transforming growth factor-b inhibits IL-4 and IFN-g production bystimulated human T cells. Int Immunol 6:469±475, 1994

Ichiki Y, Smith EA, LeRoy EC, Trojanowska M: Basic ®broblast growth factorinhibits basal and transforming growth factor-b induced collagen a2(I) geneexpression in scleroderma and normal ®broblast. J Rheumatol 24:90±95, 1997

Kasturi KN, Hatakeyama A, Murai C, Gordon R, Phelps RG, Bona CA: B cellde®ciency does not abrogate development of cutaneous hyperplasia in miceinheriting defective ®brillin-1 gene. J Autoimmun 10:505±517, 1997

Kawakami T, Ihn H, Xu W, Smith E, LeRoy C, Trojanowska M: Increasedexpression of TGF-b receptors by scleroderma ®broblast: evidence forcontribution of autocrine TGF-b signaling to scleroderma phenotype. JInvest Dermatol 110:47±51, 1998

Kulkarni AB, Huh C, Becker D, et al: Transforming growth factor-b1 null mutationin mice causes excessive in¯ammatory response and early death. Proc Natl AcadSciences USA 90:770±774, 1992

Li J, Hunter CA, Farrell JP: Anti-TGF-b treatment promotes rapid healing ofLeishmania major infection in mice by enhancing in vivo nitric oxide production.J Immunol 162:974±979, 1999

Murai S, Saito S, Kasturi KN, Bona CA: Spontaneous occurrence of anti-®brillin-1autoantibodies in tight skin mice. Autoimmunity 28:151±155, 1998

Muryoi T, Kasturi KN, Ka®na MJ, Cram DS, Harrison LC, Saki T, Bona CA:Antitopoisomerase 1 monoclonal from scleroderma patients and tight skinmouse interact with similar epitopes. J Exp Med 175:1103±1109, 1992

Nakashimia K, Yanagisawa M, Arakawa H, et al: Synergistic signaling in fetal brain bySTAT3-Smad1 complex bridged by p300. Science 284:479±482, 1999

Noben-Trauth N, Shultz LD, Brombacher F, Urban JF, Gu H, Paul WE: Aninterleukin 4 (IL-4) -independent pathway for CD4+ T cell IL-4 production isrevealed in IL-4 receptor-de®cient mice. Proc Natl Acad Sciences USA94:10838±10843, 1997

Ong C, Wong C, Roberts CR, The HS, Jirik FR: Anti-IL-4 treatment preventsdermal collagen deposition in the tight-skin mouse model of scleroderma. Eur JImmunol 28:2619±2629, 1998

Peterkofsky B, Diegelmann R: Use of a mixture of proteinase-free collagenases forthe speci®c assay of radioactive collagen in the presence of other proteins.Biochemistry 10:988±993, 1970

Phelps RG, Daian C, Shibata S, Fleischmajer R, Bona CA: Induction of skin ®brosisand autoantibodies by infusion of immunocompetent cells from tight skin miceinto C57BL/6 pa/pa mice. J Autoimmun 6:710±718, 1993

Postlethwaite AE: Role of T cells and cytokines in effective ®brosis. Int Rev Immunol12:249±259, 1995

Ridge JP, Fuchs EJ, Matzinger P: Neonatal tolerance revisited: tuning of T cells withdendritic cells. J Immunol 271:1723±1726, 1996

Rossi GA, Hunninghake GW, Gadek JE, Szapiel SU, Kawanami O, Ferrons VJ,Crystal RG: Hereditary emphysema in the tight skin mice. Am Rev Respir Dis129:850±855, 1984

Saito S, Nishimura H, Phelps RG, Wolf I, Suzuki M, Honjo T, Bona C: Inductionof skin ®brosis in mice expressing a mutated ®brillin-1 gene. Mol Med 6:825±836, 2000

Sakai LY, Keene DR, Engvall E: A new 350-kD glycoprotein is a new component ofextracellular micro®brils. J Cell Biol 103:2499±2509, 1986

Salmon-Ehr V, Serpier H, Nawrocki B, et al: Expression of interleukin-4 inscleroderma skin specimens and scleroderma ®broblast cultures. Arch Dermatol132:802±806, 1996

Schnieke A, Harbers K, Jaenisch R: Embryonic lethal mutation in mice induced byretrovirus insertion into the a1(I) collagen gene. Nature 304:315±319, 1983

Seder RA, Marth T, Sieve MC, Strober W, Letterio JJ, Roberts AB, Kelsall B:Factors involved in the differentiation of TGF-b producing cells from naiveCD4+ T cells: IL-4 and IFN-g have opposing effects, while TGF-b positivelyregulates its own production. J Immunol 160:5719±5728, 1998

Serpier H, Gillery P, Salmon-Ehr V, Garnotel R, Geroges N, Kalis B, Maquart F-X:Antagonistic effects of interferon-g and interleukin-4 on ®broblast cultures. JInvest Dermatol 109:158±162, 1997

Shero JH, Bordwell B, Roth®eld NI, Earnshae WC: High titers of autoantibodies to

142 MCGAHA ET AL THE JOURNAL OF INVESTIGATIVE DERMATOLOGY

topoisomerase 1 (ScL-70) in sera from scleroderma patients. Science 231:737±740, 1986

Shull MM, Ormsby I, Kier AB, et al: Targeted disruption of the mouse transforminggrowth factor-b1 gene results in multifocal in¯ammatory disease. Cell 359:693±698, 1992

Siracusa L, McGrath R, Ma Q, et al: A tandem duplication within the ®brillin-1 geneis associated with the mouse tight skin mutation. Genome Res 6:300±313, 1996

Siracusa LD, McGrath R, Fisher JK, Jimenez SA: The mouse tight skin (TSK)phenotype is not dependent on the presence of mature T and B lymphocytes.Mamm Genome 9:907±909, 1998

Tan FK, Arnett FC, Antohi S, et al: Autoantibodies to the extracellular matrixmicro®brillar protein, ®brillin-1, in patients with scleroderma and otherconnective tissue diseases. J Immunol 163:1066±1072, 1999

Wallace VA, Kondo S, Kono T, et al: A role for CD4+ T-cells in the pathogenesis ofskin ®brosis in tight skin mice. Eur J Immunol 24:1463±1466, 1994

White N, Needelman B, Wigley T-M, Stair RW: Interleukin-1, interleukin-2,interleukin-4, interleukin-6, tumor necrosis factor alpha and interferon gammalevels in sera from patients with scleroderma. Arthritis Rheum 35:67±92, 1992

VOL. 116, NO. 1 JANUARY 2001 AUTOIMMUNITY AND CYTOKINES IN SKIN SCLEROSIS 143