Skip to main content
Erschienen in: Arthritis Research & Therapy 3/2012

Open Access 01.06.2012 | Research article

CD109, a TGF-β co-receptor, attenuates extracellular matrix production in scleroderma skin fibroblasts

verfasst von: Xiao-Yong Man, Kenneth W Finnson, Murray Baron, Anie Philip

Erschienen in: Arthritis Research & Therapy | Ausgabe 3/2012

Abstract

Introduction

Scleroderma or systemic sclerosis (SSc) is a complex connective tissue disease characterized by fibrosis of skin and internal organs. Transforming growth factor beta (TGF-β) plays a key role in the pathogenesis of SSc fibrosis. We have previously identified CD109 as a novel TGF-β co-receptor that inhibits TGF-β signaling. The aim of the present study was to determine the role of CD109 in regulating extracellular matrix (ECM) production in human SSc skin fibroblasts.

Methods

CD109 expression was determined in skin tissue and cultured skin fibroblasts of SSc patients and normal healthy subjects, using immunofluorescence, western blot and RT-PCR. The effect of CD109 on ECM synthesis was determined by blocking CD109 expression using CD109-specific siRNA or addition of recombinant CD109 protein, and analyzing the expression of ECM components by western blot.

Results

The expression of CD109 proteinis markedly increased in SSc skin tissue in vivo and in SSc skin fibroblasts in vitro as compared to their normal counterparts. Importantly, both SSc and normal skin fibroblasts transfected with CD109-specific siRNA display increased fibronectin, collagen type I and CCN2 protein levels and enhanced Smad2/3 phosphorylation compared with control siRNA transfectants. Furthermore, addition of recombinant CD109 protein decreases TGF-β1-induced fibronectin, collagen type I and CCN2 levels in SSc and normal fibroblasts.

Conclusion

The upregulation of CD109 protein in SSc may represent an adaptation or consequence of aberrant TGF-β signaling in SSc. Our finding that CD109 is able to decrease excessive ECM production in SSc fibroblasts suggest that this molecule has potential therapeutic value for the treatment of SSc.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​ar3877) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

X-YM contributed to conception and design, acquisition of data, analysis and interpretation of data and drafting the manuscript. KWF contributed to conception and design, analysis and interpretation of data and drafting the manuscript. MB contributed to collection of samples and analysis and interpretation of data. AP contributed to conception and design, analysis and interpretation of data and drafting the manuscript. All authors contributed to revising the manuscript critically for important intellectual content, and read and approved the manuscript for publication.
Abkürzungen
bp
base pair
CCN2
connective tissue growth factor
DMEM
Dulbecco's modified Eagle's medium
ECM
extracellular matrix
GAPDH
glyceraldehyde 3-phosphate dehydrogenase
PBS
phosphate-buffered saline
PCR
polymerase chain reaction
RT
reverse transcriptase
SSc
systemic sclerosis
siRNA
small interfering RNA
TGF-β
transforming growth factor beta.

Introduction

Scleroderma or systemic sclerosis (SSc) is a complex connective tissue disorder characterized by autoimmunity, vasculopathy and progressive fibrosis of skin and internal organs [13]. SSc is commonly classified into two major clinical subsets, diffuse SSc and limited SSc, based largely on the degree of skin involvement [4]. Although there are a number of disease characteristics that differentiate between these two groups, both share the common clinical hallmark of fibrosis - characterized by excessive extracellular matrix (ECM) production, leading to disruption of normal tissue architecture and eventually organ failure [5]. Although much progress has been made in understanding the molecular mechanisms underlying the pathophysiology of SSc [2, 6, 7], there are currently no therapies to halt the fibrotic process or to slow progression of the disease [5, 8, 9].
Transforming growth factor beta (TGF-β) is a multifunctional cytokine that regulates cell proliferation, cell differentiation and ECM production [1012]. TGF-β is the most potent profibrotic cytokine known and is thought to play a key role in SSc pathogenesis [2, 6, 13, 14]. Cultured SSc fibroblasts display constitutively elevated ECM synthesis, which has been attributed to aberrant activation of autocrine TGF-β signaling [15, 16]. Some studies have demonstrated increased TGF-β receptor levels in SSc fibroblasts [1719] that might contribute to activation of autocrine TGF-β signaling [16]. However, these findings have not been universally reproduced [20, 21], emphasizing the need for further investigation.
TGF-β signaling is transduced by a pair of transmembrane serine/threonine kinases known as the TGF-β type I and type II receptors [22]. TGF-β binds the TGF-β type II receptor, which then recruits and phosphorylates the TGF-β type I receptor resulting in activation of TGF-β type I receptor kinase activity [23, 24]. The TGF-β type I receptor propagates the signal by phosphorylating intracellular Smad2 and Smad3 proteins, which form a complex with Smad4. The Smad complexes then translocate to the nucleus where they interact with various co-activators, co-repressors and transcription factors to regulate target gene expression [12, 25, 26]. Important TGF-β target genes relevant to fibrotic progression in SSc include ECM proteins such as fibronectin and collagen type I and the matricellular protein CCN2 [2, 27].
CD109 is a 180 kDa glycosylphosphatidylinositol-anchored protein belonging to the α2-macroglobulin/complement superfamily [28, 29]. Although CD109 is expressed in a variety of cell types and its expression is altered in many types of cancer, the function of this protein is poorly understood [2835]. We have recently identified CD109 as a TGF-β co-receptor and inhibitor of TGF-β signaling in human keratinocytes [36, 37]. The purpose of the current study was to determine whether CD109 expression is altered in SSc skin and whether CD109 may decrease the uncontrolled production of ECM proteins by SSc skin fibroblasts.

Materials and methods

Human subjects

Nineteen patients diagnosed with SSc (17 females, two males; Table 1) and nine normal healthy subjects (seven females and two males aged 35 to 55 years) were studied. Diagnosis of SSc was performed according to the classification criteria of the American College of Rheumatology [38]. Punch biopsies were obtained from the dorsal forearm of subjects with informed consent. Biopsies were divided into two parts, with one part placed in 10% buffered formalin for immunohistochemistry and the other part in DMEM for fibroblast isolation. This study was approved by the Institutional Review Committee of McGill University.
Table 1
Patient information
Patient
Gender
Age (years)
Modified Rodnan Skin Score
Type of systemic sclerosis
Disease duration (years)
CD109 intensity (arbitrary units)a
S1
Female
49
1
Limited
2.93
1.15
S2
Female
62
9
Limited
4.72
0.53
S3
Female
56
6
Diffuse
5.46
0.37
S4
Female
76
10
Limited
8.87
0.74
S5
Female
53
2
Limited
2.38
0.89
S6
Female
63
5
Limited
11.54
1.02
S7
Female
58
33
Diffuse
1.51
0.65
S8
Female
51
31
Diffuse
13.63
0.92
S9
Female
66
4
Limited
5.62
1.39
S10
Female
45
41
Diffuse
5.62
0.29
S11
Female
55
29
Diffuse
12.60
0.58
S12
Female
53
2
Limited
2.45
1.28
S13
Female
70
7
Diffuse
17.45
2.42
S14
Female
68
10
Diffuse
5.68
1.47
S15
Female
63
2
Limited
28.87
0.35
S16
Male
69
4
Limited
1.66
1.45
S17
Male
77
2
Limited
13.46
ND
S18
Female
49
0
Limited
6.00
ND
S19
Female
60
1
Limited
23.75
ND
Modified Rodnan Skin Score from [54]. ND, not determined. aSee Figure 2A, B.

Cell isolation and culture

Biopsies were incubated in 0.5% dispase (Invitrogen, Carlsbad, CA, USA) overnight at 4°C, and the dermis was separated from the epidermis. The epidermis was incubated with 0.25% trypsin (Invitrogen) for 20 minutes and the released keratinocytes were cultured in keratinocyte serum-free media (Invitrogen). The dermis was incubated with 0.1% collagenase (Invitrogen) overnight and the released fibroblasts were cultured in DMEM containing 10% fetal bovine serum. Experiments were performed using fibroblasts between passages 3 and 6. Fibroblasts were serum-starved for 24 hours and treated with TGF-β1 (Genzyme Corporation, Framingham, MA, USA) without or with recombinant CD109 (R&D Systems Inc., Minneapolis, MN, USA). Cell lysates were prepared and stored at -80°C until further analysis.

Immunohistochemistry

SSc and normal skin biopsies were fixed in formalin, embedded in paraffin and cut into 4 μm serial sections using a microtome. SSc and normal skin fibroblasts cultured on glass coverslips were fixed with 4% paraformaldehyde. Tissue sections and cells were deparaffinized and rehydrated, and antigen retrieval was performed by heating at 95°C for 20 minutes in sodium citrate buffer (10 mM, pH 8.5). The samples were then permeabilized with PBS containing 0.1% Triton X-100. Blocking was performed with 10% normal rabbit serum for 1 hour at room temperature. Sections were then incubated with a mouse anti-human CD109 antibody (R&D Systems Inc.) overnight at 4°C, followed by an Alexa Fluor 488-conjugated rabbit anti-mouse secondary antibody (AF-488; Invitrogen) for 2 hours at room temperature. Immunofluorescent images were obtained using a fluorescence microscope (Olympus B202; Carson Group Inc., Markham, ON, Canada) with a digital camera. Alternatively, sections incubated with the anti-human CD109 antibody as above were detected with a horseradish peroxidase-conjugated rabbit anti-mouse secondary antibody followed by diaminobenzidine staining.

Reverse transcription-polymerase chain reaction

Total RNA was extracted from SSc and normal skin fibroblasts using the RNeasy Mini Kit (Qiagen, Mississauga, ON, Canada) and reverse transcribed using MMLV reverse transcriptase with oligo-dT as primer. PCR was performed using CD109 (forward primer, 5'-GCCTTTGATTTAGATGTTGCTGTA-3'; reverse primer, 5'-TATTCCACTTTCTTCACTGTCTCG-3'; product length 188 bp) and GAPDH (forward primer, 5'-GGGGAGCCAAAAGGGTCATCATCT-3'; and reverse primer, 5'-TTGGCCAGGGGTGCTAAG-3'; product length 145 bp) primers and Taq DNA polymerase (Invitrogen). After denaturation at 95°C for 5 minutes, the reaction was performed for 25 cycles at 95°C for 30 seconds, 59°C for 30 seconds and 72°C for 30 seconds. The PCR products were separated in 1.5% agarose gel and visualized by ethidium bromide staining.
Real-time PCR was performed using the Bio-Rad CFX96 Real-Time System using the RT2 SYBR Green qPCR Master Mix (Qiagen Inc., Mississauga, ON, Canada) with CD109-specific primers (product size 147 bp, catalogue number PPH10537A; SABiosciences) and GAPDH-specific primers (forward primer, 5'-AAGATCATCAGCAATGCCTCCTG-3'; and reverse primer, 5'-TGACCTTGCCCACAGCCTT-3'; product length 228 bp). PCR was performed with an initial denaturation step for 10 minutes at 95°C followed by 40 cycles at 95°C for 15 seconds, 55°C for 30 seconds and 72°C for 30 seconds. Data were analyzed using the ΔCt method and are presented as the fold-change in ΔCt.

siRNA transfection

SSc and normal skin fibroblasts were transfected with CD109-specific siRNA (Stealth™; Invitrogen) or control siRNA using the TransIT-LT1 transfection reagent (Mirus Bio LLC, Madison, WI, USA). Cell lysates were prepared 48 hours later and were analyzed by western blot.

Western blot

Cell lysates were prepared in RIPA buffer (50 mM Tris-HCl, 1% NP-40, 0.25% sodium deoxycholate, 150 mM NaCl, 1 mM ethylenediamine tetraacetic acid, 1 mM phenylmethylsulfonyl fluoride, 1 mM Na3VO4, 1 mM NaF and Roche complete Protease Inhibitor Cocktail; Roche Applied Biosciences, Laval, QC, Canada), and protein concentrations were determined. Cell lysates (15 μg per lane) were separated by SDS-PAGE and transferred onto nitrocellulose membranes (Millipore, Bedford, MA, USA). Following blocking with 5% nonfat dry milk in Tris-buffered saline-Tween at room temperature for 1 hour, membranes were incubated overnight with antibodies against CD109 (R&D Systems Inc.), pSmad2, pSmad3 and Smad2 (all from Cell Signaling Technology, Danvers, MA, USA), collagen type I (Abcam, Cambridge, MA, USA), fibronectin (BD Biosciences, Mississauga, ON, Canada) and CCN2, Smad3 and β-actin (all from Santa Cruz Biotechnology, Santa Cruz, CA, USA) followed by incubation with a horseradish peroxidase-conjugated secondary antibody. Immunoblots were developed with enhanced-chemiluminescence reagents (GE Healthcare, Baie d'Urfe, QC, Canada). Densitometric analysis was performed using Gel-Pro Analyzer software V4.0 (Media Cybernetics, Bethesda, MD, USA) and statistical analysis was performed by one-way analysis of variance followed by an all-pairwise multiple comparison procedure (Holm-Sidak method) with an overall significance level of 0.05.

Results

CD109 protein expression is increased in SSc skin compared with normal skin in vivo

We have previously demonstrated that CD109 is expressed in vitro in normal human keratinocytes [37, 39, 40] and fibroblasts [41, 42]. Here we examined CD109 expression in SSc and normal skin tissue by immunohistochemistry. Figure 1A shows that CD109 protein is detected in SSc skin (right panel) and normal skin (left panel). Notably, the expression of CD109 protein is markedly increased in both the dermis and epidermis of SSc skin compared with those of normal skin, as detected by a more intense immunofluorescent signal using an Alexa Fluor 488-conjugated secondary antibody (Figure 1A). Similar results were obtained by immunostaining using a horseradish peroxidase-conjugated secondary antibody with diaminobenzidine as a substrate (Figure 1B).

CD109 protein expression is increased in SSc skin cells (fibroblasts and keratinocytes) compared with normal skin cells in vitro

Fibroblasts are the main cell type responsible for excessive ECM production in SSc [13], and cultured SSc fibroblasts continue to express elevated ECM production compared with normal fibroblasts [16]. We therefore analyzed CD109 expression in cultured SSc and normal skin fibroblasts, using immunofluorescence detection. The results shown in Figure 1C demonstrate that CD109 protein is detected in both SSc and normal skin fibroblasts in culture and is localized to the plasma membrane, with diffuse staining in the cytoplasm. Although the cytoplasmic staining may represent newly synthesized CD109 protein and/or CD109 protein that has undergone internalization, the method used does not allow us to precisely define the localization of CD109 in these cells. Importantly, SSc skin fibroblasts display increased CD109 protein levels compared with normal skin fibroblasts.
Because CD109 protein levels are also increased in SSc epidermis compared with normal epidermis (Figure 1A, B), we sought to determine CD109 protein expression in cultured SSc and normal keratinocytes. Figure 1D shows that CD109 protein levels are markedly increased in cell lysates from SSc keratinocytes compared with normal keratinocytes as determined by western blot (upper panel). Reprobing the membrane with an anti-β-actin antibody confirms that equal amounts of protein were loaded in each lane. Densitometric analysis of CD109 protein levels (180 kDa band) in the cell lysates of SSc keratinocytes (n = 6) and normal keratinocytes (n = 6) was then performed using β-actin as a loading control. The results indicate that SSc keratinocytes display significantly (P < 0.05) higher CD109 protein levels compared with normal keratinocytes (Figure 1E).

Elevated CD109 protein expression is not associated with an increase in CD109 mRNA expression in SSc skin fibroblasts

The above results suggest that SSc fibroblasts express higher levels of CD109 protein in vivo compared with their normal counterparts and that SSc fibroblasts continue to express elevated CD109 protein in vitro. We further examined CD109 protein expression in vitro in SSc skin fibroblasts and normal skin fibroblasts by western blot (Figure 2A). Results shown in Figure 2A demonstrate that SSc skin fibroblasts show higher CD109 protein levels compared with normal skin fibroblasts. Densitometric analysis of CD109 protein levels (180 kDa band) in the cell lysates of SSc and normal skin fibroblasts was then performed using β-actin as a loading control. Results from two independent experiments corresponding to skin fibroblasts from two different randomly selected groups of SSc patients versus normal subjects are shown in Figure 2B. Further analysis of these data obtained from 16 SSc patients (nine with limited SSc and seven with diffuse SSc) and nine normal healthy controls was performed after reorganization into normal control, limited SSc and diffuse SSc groups. One data point (outlier) from the control group (value = 1.04) was removed. Determination of the fold-change in the ratio of CD109/β-actin in the limited SSc and diffuse SSc groups compared with normal controls shows that both limited SSc and diffuse SSc skin fibroblasts display significantly (P < 0.05) higher CD109 protein levels compared with normal skin fibroblasts (Figure 2C).
We next determined CD109 mRNA expression in SSc (n = 7) and normal (n = 7) skin fibroblasts by RT-PCR. Figure 2D indicates that SSc and normal skin fibroblasts display similar levels of CD109 mRNA expression (top panel). RT-PCR of GAPDH indicates that similar amounts of GAPDH are present in all samples (Figure 2D, bottom panel). Densitometric analysis of the data shown in Figure 2D is presented in Figure 2E and indicates that CD109/GAPDH levels in SSc and normal skin fibroblasts are not significantly different (P = 0.717). We further analyzed CD109 mRNA expression levels by real-time RT-PCR in SSc (n = 4) and normal (n = 3) skin fibroblasts and obtained similar results (data not shown).

Treatment with exogenous TGF-β1does not alter CD109 protein levels in SSc and normal skin fibroblasts

TGF-β is the most potent profibrotic cytokine known and has been implicated in the pathogenesis of SSc fibrosis [2, 5]. Because our results indicate that CD109 protein levels are elevated in cultured SSc fibroblasts, we sought to determine whether TGF-β regulates CD109 protein levels. Figure 3 (top panel) shows that CD109 protein levels are higher in SSc skin fibroblasts compared with normal skin fibroblasts in the absence of TGF-β treatment (lane 1 vs. lane 7), as expected. Importantly, TGF-β treatment has no effect on CD109 protein levels in both SSc and normal skin fibroblasts (Figure 3, top panel). The responsiveness of SSc and normal fibroblasts to exogenous TGF-β is demonstrated by their increased fibronectin protein production (Figure 3, middle panel). Reprobing the membrane with an anti-β-actin antibody confirms that equal amounts of protein were loaded in each lane (Figure 3, bottom panel).

CD109 inhibits ECM and CCN2 protein production in SSc and normal skin fibroblasts

It is well documented that SSc skin fibroblasts in culture continue to express elevated levels of ECM proteins such as fibronectin and collagen type I [15]. We therefore examined whether CD109 regulates levels of these ECM proteins in SSc skin fibroblasts by blocking CD109 expression using siRNA, followed by western blot analysis. Figure 4A shows that SSc skin fibroblasts transfected with CD109-specific siRNA display a marked reduction in CD109 protein level compared with control siRNA-transfected cells, as expected (top panel). Importantly, CD109 siRNA-transfected SSc skin fibroblasts display elevated fibronectin, collagen type I and CCN2 protein levels compared with control siRNA-transfected cells (Figure 4A, middle panels). Reprobing the membrane with an anti-β-actin (Figure 4A, bottom panel) antibody confirms that equal amounts of protein were loaded in each lane. Similar results are obtained using normal skin fibroblasts (Figure 4B).

CD109 inhibits phosphorylation of Smad2 and Smad3 in SSc and normal skin fibroblasts

We have previously shown that CD109 inhibits Smad2/3 phosphorylation in human keratinocytes and other cell types [37]. Because ECM and CCN2 proteins are regulated by Smad-dependent mechanisms [27] we examined whether CD109 regulates Smad2/3 phosphorylation in SSc skin fibroblasts. Figure 4C shows that SSc skin fibroblasts transfected with CD109 siRNA display elevated Smad2 phosphorylation (top panel) and Smad3 phosphorylation (third panel) compared with control siRNA-transfected cells. Total Smad2 (second panel) and Smad3 (fourth panel) levels were not affected by CD109 siRNA knockdown (Figure 4C, middle panels). Reprobing the membrane with an anti-β-actin antibody confirms that equal amounts of protein were loaded in each lane (Figure 4C, bottom panel). Similar results were obtained using normal skin fibroblasts (Figure 4D). Although previous studies have reported elevated Smad2 and Smad3 phosphorylation in diffuse SSc fibroblasts [43, 44], our results indicate that limited SSc and normal skin fibroblasts have similar Smad2 and Smad3 phosphorylation levels (Figure 4C, D), which might be attributed to phenotypic differences in TGF-β pathway activation between limited SSc and diffuse SSc skin fibroblasts [45, 46].

Soluble recombinant CD109 protein inhibits ECM and CCN2 production in SSc fibroblasts

We have previously shown that CD109 is released from the human keratinocyte cell surface [42], indicating that endogenous CD109 exists as both membrane-tethered and soluble forms. Our previous results also demonstrate that endogenous CD109 protein released from the cell surface and soluble recombinant CD109 protein both bind TGF-β1 with high affinity and inhibit TGF-β1 binding to its signaling receptors ([42] and unpublished observations, AB and AP, 2012). We therefore examined whether soluble recombinant CD109 protein inhibits TGF-β1-induced ECM and CCN2 protein production in SSc fibroblasts. Figure 5A shows that TGF-β1 induces fibronectin, collagen type I and CCN2 protein expression in SSc and normal skin fibroblasts as expected. Importantly, addition of recombinant CD109 protein leads to a marked reduction in TGF-β1-induced fibronectin, collagen type I and CCN2 protein expression in these cells. Reprobing the membrane with an anti-β-actin antibody confirms that equal amounts of protein were loaded in each lane (Figure 5A, bottom panel). Densitometric analysis of data obtained from three different experiments confirms that addition of recombinant CD109 protein results in a significant decrease in the production of TGF-β1-induced fibronectin, collagen type I and CCN2 in limited SSc and normal skin fibroblasts (Figure 5B). The results also indicate that limited SSc skin fibroblasts display higher fibronectin and collagen type I protein levels, but similar CCN2 levels, compared with normal skin fibroblasts under basal conditions.

Discussion

SSc is a rare connective tissue disease of unknown etiology characterized by excessive ECM deposition in the skin and internal organs [2, 6, 13]. Studies over the past decade aimed at delineating the molecular mechanisms involved in SSc fibrosis have identified TGF-β as a central player and the TGF-β signaling pathway as an important target for therapeutic intervention for SSc [2, 5, 14]. We have previously identified CD109 as a TGF-β co-receptor that inhibits TGF-β signaling in human keratinocytes [37]. In the current study, we examine CD109 expression and function in limited SSc and diffuse SSc skin and normal skin in vivo, and in SSc and normal dermal fibroblasts and epidermal keratinocytes in vitro. Our results show that CD109 protein levels are elevated in vivo in both the dermis and epidermis of SSc skin compared with normal skin and in vitro in SSc dermal fibroblasts and epidermal keratinocytes compared with normal fibroblasts and keratinocytes, respectively. Although cultured SSc skin fibroblasts show higher CD109 protein levels compared with normal skin fibroblasts, they do not appear to display elevated CD109 mRNA levels. Elevated CD109 protein levels were observed in the skin tissue and fibroblasts from patients with both limited SSc and diffuse SSc, suggesting that increased CD109 protein expression might be a general feature of SSc. We also demonstrate that treatment with exogenous TGF-β1 does not alter CD109 protein levels in SSc or normal skin fibroblasts, suggesting that CD109 is not a direct target of TGF-β in these cells. Moreover, we found that CD109 inhibits ECM (fibronectin and collagen type I) and CCN2 protein production in both SSc and normal skin fibroblasts - as evidenced by the findings that CD109 siRNA-transfected SSc and normal skin fibroblasts display elevated fibronectin, collagen type I and CCN2 protein levels, whereas treatment with a recombinant CD109 protein leads to a decrease in levels of these proteins. In addition, we show that CD109 inhibits phosphorylation of Smad2 and Smad3 in SSc and normal fibroblasts. Taken together, this study identifies CD109 as a novel protein overexpressed in limited SSc and diffuse SSc skin fibroblasts and indicates that CD109 inhibits Smad2/3 signaling and ECM/CCN2 production in these cells, as in normal skin fibroblasts.
TGF-β co-receptors including endoglin, betaglycan and CD109 are important regulators of TGF-β signaling, and recent evidence indicates that they may modulate fibrotic gene expression in SSc fibroblasts. Endoglin expression has been reported to be higher in diffuse SSc skin fibroblasts compared with normal fibroblasts, and endoglin expression levels were shown to increase with disease progression [47]. A more recent study demonstrated that endoglin levels correlate with profibrotic marker (collagen I and CCN2) levels in diffuse SSc skin fibroblasts and that endoglin promotes profibrogenic Smad1 signaling in these cells [48]. In addition, cell surface endoglin and betaglycan expression levels were shown to be increased in diffuse SSc skin fibroblasts compared with normal fibroblasts and overexpression of betaglycan was sufficient to promote basal and TGF-β-induced CCN2 promoter activity in a mouse fibroblast cell line (NIH3T3) [49]. Our results showing that CD109 protein levels are elevated in both limited SSc and diffuse SSc skin tissue and cultured fibroblasts compared with controls suggest that upregulation of CD109 in these SSc subtypes may involve a common pathophysiological mechanism. Further studies to determine the expression and function of TGF-β co-receptors in the different clinical subsets of SSc, in particular their relevance to TGF-β-driven profibrogenic Smad1 signaling, is an important avenue for future research.
An important question raised from our results is why CD109 protein levels but not mRNA levels are increased in SSc fibroblasts. Our data showing that the levels of CD109 protein, but not of mRNA, are higher in limited SSc and diffuse SSc skin fibroblasts compared with normal skin fibroblasts suggests that post-transcriptional regulation of CD109 may be altered in limited SSc and diffuse SSc skin. Such alteration may involve enhanced CD109 protein synthesis and/or decreased CD109 protein degradation. Our preliminary results demonstrating that CD109 protein degradation is impaired in SSc fibroblasts (AB and AP, 2012, unpublished results) suggest that impaired CD109 degradation may be responsible for the elevated CD109 protein levels in SSc fibroblasts. It is interesting to note in this regard that several studies have reported increased TGF-β receptor protein levels in SSc fibroblasts [21, 50], leading to the suggestion that TGF-β receptor stability is increased in SSc due to impairment of TGF-β receptor degradation [44]. Furthermore, recent microarray data [46, 51] support our finding that CD109 mRNA levels in SSc and normal fibroblasts are similar. However, quantitative PCR analysis of RNA from a higher number of limited SSc and diffuse SSc dermal fibroblasts with age-matched, sex-matched and gender-matched control samples will be required to confirm these results.
Paradoxically, despite the elevated CD109 protein levels in SSc skin fibroblasts and the demonstrated ability of endogenous CD109 protein to inhibit ECM and CCN2 production, the levels of ECM and CCN2 in SSc fibroblasts remain elevated. One possible explanation for these results is that the upregulation of CD109 in SSc fibroblasts is not sufficient to completely counteract the activation of TGF-β or other profibrotic pathways in SSc. This notion is supported by our finding that addition of a recombinant CD109 protein to SSc fibroblasts, which already express elevated CD109 protein levels, is still able to decrease ECM and CCN2 levels. Whether the upregulation of CD109 is an adaptive response or a consequence of aberrant TGF-β signaling in SSc remains to be determined. Our results showing that CD109 is not a direct target of TGF-β in SSc and normal skin fibroblasts suggests that CD109 upregulation in SSc fibroblasts may involve TGF-β-independent mechanisms.
The most promising finding in the current study is that, despite the already upregulated CD109 protein expression in SSc, the addition of further exogenous CD109 is able to downregulate ECM production. Aberrant activation of autocrine TGF-β signaling is a critical factor involved in the maintenance of the fibrotic phenotype of SSc fibroblasts [16]. Identifying factors that inhibit TGF-β signaling in fibroblasts may thus lead to the development of novel anti-TGF-β therapies for the treatment of SSc. Our results showing that CD109 siRNA transfection leads to a marked increase, and that addition of recombinant CD109 protein results in a significant decrease, in the production of fibronectin, collagen type I and CCN2 in both SSc and normal skin fibroblasts suggest that CD109 has potent antifibrotic effects in these cells. In addition, our finding that CD109 siRNA knockdown in SSc and normal skin fibroblasts is associated with enhanced Smad2/3 phosphorylation suggests that endogenous CD109 inhibits autocrine TGF-β signaling in these cells. These results suggest that mechanism by which CD109 decreases ECM and CCN2 production in SSc and normal skin fibroblasts may involve inhibition of Smad2/3 phosphorylation. Together, our results suggest that recombinant CD109 protein is a promising candidate for antifibrotic therapy in SSc.
In addition to playing a possible pathophysiological role in SSc, the elevated CD109 protein levels may serve as a biomarker of disease activity in SSc. CD109 is a member of the α2-macroglobulin/complement family of thioester-containing proteins and has been shown to be expressed in endothelial cells, platelets and activated T cells [28, 29]. Recent studies have shown that CD109 is differentially expressed in a variety of human tumors [3034, 37, 52, 53], leading to the suggestion that CD109 may represent a novel biomarker for certain cancers. The relatively small number of samples examined in the current study does not allow us to correlate CD109 expression levels with disease severity and/or duration. More work is needed to determine whether CD109 levels are differentially expressed at specific phases of disease activity in limited SSC and diffuse SSc.

Conclusions

Despite the findings that CD109 is upregulated in SSc skin and cultured SSc skin fibroblasts, and that blocking CD109 expression in SSc cells leads to further production of ECM, it is clear that the CD109 response in SSc is insufficient to inhibit the production of excess ECM. Importantly, however, exposure of SSc fibroblasts to additional exogenous CD109 does result in the downregulation of excess ECM production - it is therefore worthwhile pursuing the role of CD109 as an antifibrotic agent and exploring whether CD109 protein expression represents a biomarker for active disease in SSc.

Acknowledgements

The authors would like to express their appreciation to D Dicapua and Dr L Lessard for assistance with collection of the skin samples. This work was supported by grants from the Canadian Institutes of Health Research Operating (to AP), the Canadian Institutes of Health Research Strategic Training Initiative (to MB and AP) and the Canadian Institutes of Health Research New Emerging Team (to AP and MB). X-YM was supported by the Canadian Institutes of Health Research Training Initiative grant.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

X-YM contributed to conception and design, acquisition of data, analysis and interpretation of data and drafting the manuscript. KWF contributed to conception and design, analysis and interpretation of data and drafting the manuscript. MB contributed to collection of samples and analysis and interpretation of data. AP contributed to conception and design, analysis and interpretation of data and drafting the manuscript. All authors contributed to revising the manuscript critically for important intellectual content, and read and approved the manuscript for publication.
Literatur
1.
Zurück zum Zitat Wick G, Backovic A, Rabensteiner E, Plank N, Schwendtner C, Sgonc R: The immunology of fibrosis: innate and adaptive responses. Trends Immunol. 2010, 31: 110-119. 10.1016/j.it.2009.12.001.PubMedCentralCrossRefPubMed Wick G, Backovic A, Rabensteiner E, Plank N, Schwendtner C, Sgonc R: The immunology of fibrosis: innate and adaptive responses. Trends Immunol. 2010, 31: 110-119. 10.1016/j.it.2009.12.001.PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Rosenbloom J, Castro SV, Jimenez SA: Narrative review: fibrotic diseases: cellular and molecular mechanisms and novel therapies. Ann Intern Med. 2010, 152: 159-166.CrossRefPubMed Rosenbloom J, Castro SV, Jimenez SA: Narrative review: fibrotic diseases: cellular and molecular mechanisms and novel therapies. Ann Intern Med. 2010, 152: 159-166.CrossRefPubMed
3.
Zurück zum Zitat Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M: Mechanisms in the loss of capillaries in systemic sclerosis: angiogenesis versus vasculogenesis. J Cell Mol Med. 2010, 14: 1241-1254. 10.1111/j.1582-4934.2010.01027.x.PubMedCentralCrossRefPubMed Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M: Mechanisms in the loss of capillaries in systemic sclerosis: angiogenesis versus vasculogenesis. J Cell Mol Med. 2010, 14: 1241-1254. 10.1111/j.1582-4934.2010.01027.x.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Hachulla E, Launay D: Diagnosis and classification of systemic sclerosis. Clin Rev Allergy Immunol. 2011, 40: 78-83. 10.1007/s12016-010-8198-y.CrossRefPubMed Hachulla E, Launay D: Diagnosis and classification of systemic sclerosis. Clin Rev Allergy Immunol. 2011, 40: 78-83. 10.1007/s12016-010-8198-y.CrossRefPubMed
5.
Zurück zum Zitat Varga J, Pasche B: Transforming growth factor β as a therapeutic target in systemic sclerosis. Nat Rev Rheumatol. 2009, 5: 200-206. 10.1038/nrrheum.2009.26.PubMedCentralCrossRefPubMed Varga J, Pasche B: Transforming growth factor β as a therapeutic target in systemic sclerosis. Nat Rev Rheumatol. 2009, 5: 200-206. 10.1038/nrrheum.2009.26.PubMedCentralCrossRefPubMed
6.
Zurück zum Zitat Szodoray P, Kiss E: Progressive systemic sclerosis - from the molecular background to innovative therapies. Front Biosci (Elite Ed). 2010, 2: 521-525.CrossRef Szodoray P, Kiss E: Progressive systemic sclerosis - from the molecular background to innovative therapies. Front Biosci (Elite Ed). 2010, 2: 521-525.CrossRef
7.
Zurück zum Zitat Smith G, Chan E: Molecular pathogenesis of skin fibrosis: insight from animal models. Curr Rheumatol Rep. 2010, 12: 26-33. 10.1007/s11926-009-0080-7.PubMedCentralCrossRefPubMed Smith G, Chan E: Molecular pathogenesis of skin fibrosis: insight from animal models. Curr Rheumatol Rep. 2010, 12: 26-33. 10.1007/s11926-009-0080-7.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Postlethwaite AE, Harris LJ, Raza SH, Kodura S, Akhigbe T: Pharmacotherapy of systemic sclerosis. Expert Opin Pharmacother. 2010, 11: 789-806. 10.1517/14656561003592177.PubMedCentralCrossRefPubMed Postlethwaite AE, Harris LJ, Raza SH, Kodura S, Akhigbe T: Pharmacotherapy of systemic sclerosis. Expert Opin Pharmacother. 2010, 11: 789-806. 10.1517/14656561003592177.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Asano Y: Future treatments in systemic sclerosis. J Dermatol. 2010, 37: 54-70. 10.1111/j.1346-8138.2009.00758.x.CrossRefPubMed Asano Y: Future treatments in systemic sclerosis. J Dermatol. 2010, 37: 54-70. 10.1111/j.1346-8138.2009.00758.x.CrossRefPubMed
10.
Zurück zum Zitat Wu MY, Hill CS: TGF-β superfamily signaling in embryonic development and homeostasis. Dev Cell. 2009, 16: 329-343. 10.1016/j.devcel.2009.02.012.CrossRefPubMed Wu MY, Hill CS: TGF-β superfamily signaling in embryonic development and homeostasis. Dev Cell. 2009, 16: 329-343. 10.1016/j.devcel.2009.02.012.CrossRefPubMed
11.
Zurück zum Zitat Wharton K, Derynck R: TGF-β family signaling: novel insights in development and disease. Development. 2009, 136: 3691-3697. 10.1242/dev.040584.CrossRefPubMed Wharton K, Derynck R: TGF-β family signaling: novel insights in development and disease. Development. 2009, 136: 3691-3697. 10.1242/dev.040584.CrossRefPubMed
12.
Zurück zum Zitat Moustakas A, Heldin CH: The regulation of TGFβ signal transduction. Development. 2009, 136: 3699-3714. 10.1242/dev.030338.CrossRefPubMed Moustakas A, Heldin CH: The regulation of TGFβ signal transduction. Development. 2009, 136: 3699-3714. 10.1242/dev.030338.CrossRefPubMed
13.
Zurück zum Zitat Jinnin M: Mechanisms of skin fibrosis in systemic sclerosis. J Dermatol. 2010, 37: 11-25. 10.1111/j.1346-8138.2009.00738.x.CrossRefPubMed Jinnin M: Mechanisms of skin fibrosis in systemic sclerosis. J Dermatol. 2010, 37: 11-25. 10.1111/j.1346-8138.2009.00738.x.CrossRefPubMed
14.
Zurück zum Zitat Varga J, Whitfield ML: Transforming growth factor-β in systemic sclerosis (scleroderma). Front Biosci (Schol Ed). 2009, 1: 226-235.CrossRef Varga J, Whitfield ML: Transforming growth factor-β in systemic sclerosis (scleroderma). Front Biosci (Schol Ed). 2009, 1: 226-235.CrossRef
15.
Zurück zum Zitat Ihn H: Autocrine TGF-β signaling in the pathogenesis of systemic sclerosis. J Dermatol Sci. 2008, 49: 103-113. 10.1016/j.jdermsci.2007.05.014.CrossRefPubMed Ihn H: Autocrine TGF-β signaling in the pathogenesis of systemic sclerosis. J Dermatol Sci. 2008, 49: 103-113. 10.1016/j.jdermsci.2007.05.014.CrossRefPubMed
16.
Zurück zum Zitat Pannu J, Trojanowska M: Recent advances in fibroblast signaling and biology in scleroderma. Curr Opin Rheumatol. 2004, 16: 739-745. 10.1097/01.bor.0000137894.63091.1a.CrossRefPubMed Pannu J, Trojanowska M: Recent advances in fibroblast signaling and biology in scleroderma. Curr Opin Rheumatol. 2004, 16: 739-745. 10.1097/01.bor.0000137894.63091.1a.CrossRefPubMed
17.
Zurück zum Zitat Ihn H, Yamane K, Kubo M, Tamaki K: Blockade of endogenous transforming growth factor β signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with increased expression of transforming growth factor β receptors. Arthritis Rheum. 2001, 44: 474-480. 10.1002/1529-0131(200102)44:2<474::AID-ANR67>3.0.CO;2-#.CrossRefPubMed Ihn H, Yamane K, Kubo M, Tamaki K: Blockade of endogenous transforming growth factor β signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with increased expression of transforming growth factor β receptors. Arthritis Rheum. 2001, 44: 474-480. 10.1002/1529-0131(200102)44:2<474::AID-ANR67>3.0.CO;2-#.CrossRefPubMed
18.
Zurück zum Zitat Kawakami T, Ihn H, Xu W, Smith E, LeRoy C, Trojanowska M: Increased expression of TGF-β receptors by scleroderma fibroblasts: evidence for contribution of autocrine TGF-β signaling to scleroderma phenotype. J Invest Dermatol. 1998, 110: 47-51. 10.1046/j.1523-1747.1998.00073.x.CrossRefPubMed Kawakami T, Ihn H, Xu W, Smith E, LeRoy C, Trojanowska M: Increased expression of TGF-β receptors by scleroderma fibroblasts: evidence for contribution of autocrine TGF-β signaling to scleroderma phenotype. J Invest Dermatol. 1998, 110: 47-51. 10.1046/j.1523-1747.1998.00073.x.CrossRefPubMed
19.
Zurück zum Zitat Kubo M, Ihn H, Yamane K, Tamaki K: Upregulated expression of transforming growth factor-β receptors in dermal fibroblasts of skin sections from patients with systemic sclerosis. J Rheumatol. 2002, 29: 2558-2564.PubMed Kubo M, Ihn H, Yamane K, Tamaki K: Upregulated expression of transforming growth factor-β receptors in dermal fibroblasts of skin sections from patients with systemic sclerosis. J Rheumatol. 2002, 29: 2558-2564.PubMed
20.
Zurück zum Zitat Dong C, Zhu S, Wang T, Yoon W, Li Z, Alvarez RJ, ten Dijke P, White B, Wigley FM, Goldschmidt-Clermont PJ: Deficient Smad7 expression: a putative molecular defect in scleroderma. Proc Natl Acad Sci USA. 2002, 99: 3908-3913. 10.1073/pnas.062010399.PubMedCentralCrossRefPubMed Dong C, Zhu S, Wang T, Yoon W, Li Z, Alvarez RJ, ten Dijke P, White B, Wigley FM, Goldschmidt-Clermont PJ: Deficient Smad7 expression: a putative molecular defect in scleroderma. Proc Natl Acad Sci USA. 2002, 99: 3908-3913. 10.1073/pnas.062010399.PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Pannu J, Gore-Hyer E, Yamanaka M, Smith EA, Rubinchik S, Dong JY, Jablonska S, Blaszczyk M, Trojanowska M: An increased transforming growth factor β receptor type I:type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor β receptor type II in scleroderma. Arthritis Rheum. 2004, 50: 1566-1577. 10.1002/art.20225.CrossRefPubMed Pannu J, Gore-Hyer E, Yamanaka M, Smith EA, Rubinchik S, Dong JY, Jablonska S, Blaszczyk M, Trojanowska M: An increased transforming growth factor β receptor type I:type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor β receptor type II in scleroderma. Arthritis Rheum. 2004, 50: 1566-1577. 10.1002/art.20225.CrossRefPubMed
22.
Zurück zum Zitat Thatcher JD: The TGF-β signal transduction pathway. Sci Signal. 2010, 3: tr4-10.1126/scisignal.3119tr4.PubMed Thatcher JD: The TGF-β signal transduction pathway. Sci Signal. 2010, 3: tr4-10.1126/scisignal.3119tr4.PubMed
23.
Zurück zum Zitat Massague J, Gomis RR: The logic of TGF-β signaling. FEBS Lett. 2006, 580: 2811-2820. 10.1016/j.febslet.2006.04.033.CrossRefPubMed Massague J, Gomis RR: The logic of TGF-β signaling. FEBS Lett. 2006, 580: 2811-2820. 10.1016/j.febslet.2006.04.033.CrossRefPubMed
24.
Zurück zum Zitat Shi Y, Massague J: Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell. 2003, 113: 685-700. 10.1016/S0092-8674(03)00432-X.CrossRefPubMed Shi Y, Massague J: Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell. 2003, 113: 685-700. 10.1016/S0092-8674(03)00432-X.CrossRefPubMed
25.
Zurück zum Zitat Schmierer B, Hill CS: TGFβ-Smad signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol. 2007, 8: 970-982. 10.1038/nrm2297.CrossRefPubMed Schmierer B, Hill CS: TGFβ-Smad signal transduction: molecular specificity and functional flexibility. Nat Rev Mol Cell Biol. 2007, 8: 970-982. 10.1038/nrm2297.CrossRefPubMed
26.
Zurück zum Zitat Ross S, Hill CS: How the Smads regulate transcription. Int J Biochem Cell Biol. 2008, 40: 383-408. 10.1016/j.biocel.2007.09.006.CrossRefPubMed Ross S, Hill CS: How the Smads regulate transcription. Int J Biochem Cell Biol. 2008, 40: 383-408. 10.1016/j.biocel.2007.09.006.CrossRefPubMed
27.
Zurück zum Zitat Leask A: Targeting the TGFβ, endothelin-1 and CCN2 axis to combat fibrosis in scleroderma. Cell Signal. 2008, 20: 1409-1414. 10.1016/j.cellsig.2008.01.006.CrossRefPubMed Leask A: Targeting the TGFβ, endothelin-1 and CCN2 axis to combat fibrosis in scleroderma. Cell Signal. 2008, 20: 1409-1414. 10.1016/j.cellsig.2008.01.006.CrossRefPubMed
28.
Zurück zum Zitat Lin M, Sutherland DR, Horsfall W, Totty N, Yeo E, Nayar R, Wu X-F, Schuh AC: Cell surface antigen CD109 is a novel member of the alpha 2 macroglobulin/C3, C4, C5 family of thioester-containing proteins. Blood. 2002, 99: 1683-1691. 10.1182/blood.V99.5.1683.CrossRefPubMed Lin M, Sutherland DR, Horsfall W, Totty N, Yeo E, Nayar R, Wu X-F, Schuh AC: Cell surface antigen CD109 is a novel member of the alpha 2 macroglobulin/C3, C4, C5 family of thioester-containing proteins. Blood. 2002, 99: 1683-1691. 10.1182/blood.V99.5.1683.CrossRefPubMed
29.
Zurück zum Zitat Solomon KR, Sharma P, Chan M, Morrison PT, Finberg RW: CD109 represents a novel branch of the α2-macroglobulin/complement gene family. Gene. 2004, 327: 171-183. 10.1016/j.gene.2003.11.025.CrossRefPubMed Solomon KR, Sharma P, Chan M, Morrison PT, Finberg RW: CD109 represents a novel branch of the α2-macroglobulin/complement gene family. Gene. 2004, 327: 171-183. 10.1016/j.gene.2003.11.025.CrossRefPubMed
30.
Zurück zum Zitat Hagiwara S, Murakumo Y, Mii S, Shigetomi T, Yamamoto N, Furue H, Ueda M, Takahashi M: Processing of CD109 by furin and its role in the regulation of TGF-β signaling. Oncogene. 2010, 29: 2181-2191. 10.1038/onc.2009.506.CrossRefPubMed Hagiwara S, Murakumo Y, Mii S, Shigetomi T, Yamamoto N, Furue H, Ueda M, Takahashi M: Processing of CD109 by furin and its role in the regulation of TGF-β signaling. Oncogene. 2010, 29: 2181-2191. 10.1038/onc.2009.506.CrossRefPubMed
31.
Zurück zum Zitat Ohshima Y, Yajima I, Kumasaka MY, Yanagishita T, Watanabe D, Takahashi M, Inoue Y, Ihn H, Matsumoto Y, Kato M: CD109 expression levels in malignant melanoma. J Dermatol Sci. 2009, 57: 140-142.CrossRef Ohshima Y, Yajima I, Kumasaka MY, Yanagishita T, Watanabe D, Takahashi M, Inoue Y, Ihn H, Matsumoto Y, Kato M: CD109 expression levels in malignant melanoma. J Dermatol Sci. 2009, 57: 140-142.CrossRef
32.
Zurück zum Zitat Hockla A, Radisky DC, Radisky ES: Mesotrypsin promotes malignant growth of breast cancer cells through shedding of CD109. Breast Cancer Res Treat. 2009, 124: 27-38.PubMedCentralCrossRefPubMed Hockla A, Radisky DC, Radisky ES: Mesotrypsin promotes malignant growth of breast cancer cells through shedding of CD109. Breast Cancer Res Treat. 2009, 124: 27-38.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Hasegawa M, Moritani S, Murakumo Y, Sato T, Hagiwara S, Suzuki C, Mii S, Jijiwa M, Enomoto A, Asai N, Ishihara S, Takahashi M: CD109 expression in basal-like breast carcinoma. Pathol Int. 2008, 58: 288-294. 10.1111/j.1440-1827.2008.02225.x.CrossRefPubMed Hasegawa M, Moritani S, Murakumo Y, Sato T, Hagiwara S, Suzuki C, Mii S, Jijiwa M, Enomoto A, Asai N, Ishihara S, Takahashi M: CD109 expression in basal-like breast carcinoma. Pathol Int. 2008, 58: 288-294. 10.1111/j.1440-1827.2008.02225.x.CrossRefPubMed
34.
Zurück zum Zitat Hagiwara S, Murakumo Y, Sato T, Shigetom T, Mitsudo K, Tohnai I, Ueda M, Takahashi M: Up-regulation of CD109 expression is associated with carcinogenesis of the squamous epithelium of the oral cavity. Cancer Sci. 2008, 99: 1916-1923.CrossRefPubMed Hagiwara S, Murakumo Y, Sato T, Shigetom T, Mitsudo K, Tohnai I, Ueda M, Takahashi M: Up-regulation of CD109 expression is associated with carcinogenesis of the squamous epithelium of the oral cavity. Cancer Sci. 2008, 99: 1916-1923.CrossRefPubMed
35.
Zurück zum Zitat Hasegawa M, Hagiwara S, Sato T, Jijiwa M, Murakumo Y, Maeda M, Moritani S, Ichihara S, Takahashi M: CD109, a new marker for myoepithelial cells of mammary, salivary, and lacrimal glands and prostate basal cells. Pathol Int. 2007, 57: 245-250. 10.1111/j.1440-1827.2007.02097.x.CrossRefPubMed Hasegawa M, Hagiwara S, Sato T, Jijiwa M, Murakumo Y, Maeda M, Moritani S, Ichihara S, Takahashi M: CD109, a new marker for myoepithelial cells of mammary, salivary, and lacrimal glands and prostate basal cells. Pathol Int. 2007, 57: 245-250. 10.1111/j.1440-1827.2007.02097.x.CrossRefPubMed
36.
Zurück zum Zitat Bizet AA, Liu K, Tran-Khanh N, Saksena A, Vorstenbosch J, Finnson KW, Buschmann MD, Philip A: The TGF-β co-receptor, CD109, promotes internalization and degradation of TGF-β receptors. Biochim Biophys Acta Mol Cell Res. 2011, 1813: 742-753. 10.1016/j.bbamcr.2011.01.028.CrossRef Bizet AA, Liu K, Tran-Khanh N, Saksena A, Vorstenbosch J, Finnson KW, Buschmann MD, Philip A: The TGF-β co-receptor, CD109, promotes internalization and degradation of TGF-β receptors. Biochim Biophys Acta Mol Cell Res. 2011, 1813: 742-753. 10.1016/j.bbamcr.2011.01.028.CrossRef
37.
Zurück zum Zitat Finnson KW, Tam BY, Liu K, Marcoux A, Lepage P, Roy S, Bizet AA, Philip A: Identification of CD109 as part of the TGF-β receptor system in human keratinocytes. FASEB J. 2006, 20: 1525-1527. 10.1096/fj.05-5229fje.CrossRefPubMed Finnson KW, Tam BY, Liu K, Marcoux A, Lepage P, Roy S, Bizet AA, Philip A: Identification of CD109 as part of the TGF-β receptor system in human keratinocytes. FASEB J. 2006, 20: 1525-1527. 10.1096/fj.05-5229fje.CrossRefPubMed
38.
Zurück zum Zitat Lonzetti LS, Joyal F, Raynauld JP, Roussin A, Goulet JR, Rich E, Choquette D, Raymond Y, Senecal JL: Updating the American College of Rheumatology preliminary classification criteria for systemic sclerosis: addition of severe nailfold capillaroscopy abnormalities markedly increases the sensitivity for limited scleroderma. Arthritis Rheum. 2001, 44: 735-736. 10.1002/1529-0131(200103)44:3<735::AID-ANR125>3.0.CO;2-F.CrossRefPubMed Lonzetti LS, Joyal F, Raynauld JP, Roussin A, Goulet JR, Rich E, Choquette D, Raymond Y, Senecal JL: Updating the American College of Rheumatology preliminary classification criteria for systemic sclerosis: addition of severe nailfold capillaroscopy abnormalities markedly increases the sensitivity for limited scleroderma. Arthritis Rheum. 2001, 44: 735-736. 10.1002/1529-0131(200103)44:3<735::AID-ANR125>3.0.CO;2-F.CrossRefPubMed
39.
Zurück zum Zitat Tam B, Germain L, Philip A: TGF-β receptor expression on human keratinocytes: a 150 kDa GPI-anchored TGF-β1 binding protein forms a heteromeric complex with type I and type II receptors. J Cell Biochem. 1998, 70: 573-586. 10.1002/(SICI)1097-4644(19980915)70:4<573::AID-JCB13>3.0.CO;2-I.CrossRefPubMed Tam B, Germain L, Philip A: TGF-β receptor expression on human keratinocytes: a 150 kDa GPI-anchored TGF-β1 binding protein forms a heteromeric complex with type I and type II receptors. J Cell Biochem. 1998, 70: 573-586. 10.1002/(SICI)1097-4644(19980915)70:4<573::AID-JCB13>3.0.CO;2-I.CrossRefPubMed
40.
Zurück zum Zitat Tam BYY, Finnson KW, Philip A: Glycosylphosphatidylinositol-anchored proteins regulate transforming growth factor-β signaling in human keratinocytes. J Biol Chem. 2003, 278: 49610-49617. 10.1074/jbc.M308492200.CrossRefPubMed Tam BYY, Finnson KW, Philip A: Glycosylphosphatidylinositol-anchored proteins regulate transforming growth factor-β signaling in human keratinocytes. J Biol Chem. 2003, 278: 49610-49617. 10.1074/jbc.M308492200.CrossRefPubMed
41.
Zurück zum Zitat Tam B, Philip A: Transforming growth factor-β receptor expression on human skin fibroblasts: dimeric complex formation of type I and type II receptors and identification of glycosyl phosphatidylinositol-anchored transforming growth factor-β binding proteins. J Cell Physiol. 1998, 176: 553-564. 10.1002/(SICI)1097-4652(199809)176:3<553::AID-JCP12>3.0.CO;2-0.CrossRefPubMed Tam B, Philip A: Transforming growth factor-β receptor expression on human skin fibroblasts: dimeric complex formation of type I and type II receptors and identification of glycosyl phosphatidylinositol-anchored transforming growth factor-β binding proteins. J Cell Physiol. 1998, 176: 553-564. 10.1002/(SICI)1097-4652(199809)176:3<553::AID-JCP12>3.0.CO;2-0.CrossRefPubMed
42.
Zurück zum Zitat Tam B, Larouche D, Germain L, Hooper N, Philip A: Characterization of a 150 kDa accessory receptor for TGF-β1 on keratinocytes: direct evidence for a GPI anchor and ligand binding of the released form. J Cell Biochem. 2001, 83: 494-507. 10.1002/jcb.1074.CrossRefPubMed Tam B, Larouche D, Germain L, Hooper N, Philip A: Characterization of a 150 kDa accessory receptor for TGF-β1 on keratinocytes: direct evidence for a GPI anchor and ligand binding of the released form. J Cell Biochem. 2001, 83: 494-507. 10.1002/jcb.1074.CrossRefPubMed
43.
Zurück zum Zitat Ihn H, Yamane K, Asano Y, Jinnin M, Tamaki K: Constitutively phosphorylated Smad3 interacts with Sp1 and p300 in scleroderma fibroblasts. Rheumatology (Oxford). 2006, 45: 157-165.CrossRef Ihn H, Yamane K, Asano Y, Jinnin M, Tamaki K: Constitutively phosphorylated Smad3 interacts with Sp1 and p300 in scleroderma fibroblasts. Rheumatology (Oxford). 2006, 45: 157-165.CrossRef
44.
Zurück zum Zitat Asano Y, Ihn H, Yamane K, Kubo M, Tamaki K: Impaired Smad7-Smurf-mediated negative regulation of TGF-β signaling in scleroderma fibroblasts. J Clin Invest. 2004, 113: 253-264.PubMedCentralCrossRefPubMed Asano Y, Ihn H, Yamane K, Kubo M, Tamaki K: Impaired Smad7-Smurf-mediated negative regulation of TGF-β signaling in scleroderma fibroblasts. J Clin Invest. 2004, 113: 253-264.PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Sargent JL, Milano A, Bhattacharyya S, Varga J, Connolly MK, Chang HY, Whitfield ML: A TGFβ-responsive gene signature is associated with a subset of diffuse scleroderma with increased disease severity. J Invest Dermatol. 2010, 130: 694-705. 10.1038/jid.2009.318.CrossRefPubMed Sargent JL, Milano A, Bhattacharyya S, Varga J, Connolly MK, Chang HY, Whitfield ML: A TGFβ-responsive gene signature is associated with a subset of diffuse scleroderma with increased disease severity. J Invest Dermatol. 2010, 130: 694-705. 10.1038/jid.2009.318.CrossRefPubMed
46.
Zurück zum Zitat Milano A, Pendergrass SA, Sargent JL, George LK, McCalmont TH, Connolly MK, Whitfield ML: Molecular subsets in the gene expression signatures of scleroderma skin. PLoS ONE. 2008, 3: e2696-10.1371/journal.pone.0002696.PubMedCentralCrossRefPubMed Milano A, Pendergrass SA, Sargent JL, George LK, McCalmont TH, Connolly MK, Whitfield ML: Molecular subsets in the gene expression signatures of scleroderma skin. PLoS ONE. 2008, 3: e2696-10.1371/journal.pone.0002696.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Leask A, Abraham DJ, Finlay DR, Holmes A, Pennington D, Shi-Wen X, Chen Y, Venstrom K, Dou X, Ponticos M, Black C, Bernabeu C, Jackman JK, Findell PR, Conolly MK: Dysregulation of transforming growth factor β signaling in scleroderma: overexpression of endoglin in cutaneous scleroderma fibroblasts. Arthritis Rheum. 2002, 46: 1857-1865. 10.1002/art.10333.CrossRefPubMed Leask A, Abraham DJ, Finlay DR, Holmes A, Pennington D, Shi-Wen X, Chen Y, Venstrom K, Dou X, Ponticos M, Black C, Bernabeu C, Jackman JK, Findell PR, Conolly MK: Dysregulation of transforming growth factor β signaling in scleroderma: overexpression of endoglin in cutaneous scleroderma fibroblasts. Arthritis Rheum. 2002, 46: 1857-1865. 10.1002/art.10333.CrossRefPubMed
48.
Zurück zum Zitat Morris E, Chrobak I, Bujor A, Hant F, Mummery C, Ten Dijke P, Trojanowska M: Endoglin promotes TGF-β/Smad1 signaling in scleroderma fibroblasts. J Cell Physiol. 2011, 226: 3340-3348. 10.1002/jcp.22690.PubMedCentralCrossRefPubMed Morris E, Chrobak I, Bujor A, Hant F, Mummery C, Ten Dijke P, Trojanowska M: Endoglin promotes TGF-β/Smad1 signaling in scleroderma fibroblasts. J Cell Physiol. 2011, 226: 3340-3348. 10.1002/jcp.22690.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Holmes AM, Ponticos M, Shi-Wen X, Denton CP, Abraham DJ: Elevated CCN2 expression in scleroderma: a putative role for the TGF-β accessory receptors TGFβRIII and endoglin. J Cell Commun Signal. 2011, 5: 173-177. 10.1007/s12079-011-0140-4.PubMedCentralCrossRefPubMed Holmes AM, Ponticos M, Shi-Wen X, Denton CP, Abraham DJ: Elevated CCN2 expression in scleroderma: a putative role for the TGF-β accessory receptors TGFβRIII and endoglin. J Cell Commun Signal. 2011, 5: 173-177. 10.1007/s12079-011-0140-4.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Kubo M, Ihn H, Yamane K, Tamaki K: Up-regulated expression of transforming growth factor β receptors in dermal fibroblasts in skin sections from patients with localized scleroderma. Arthritis Rheum. 2001, 44: 731-734. 10.1002/1529-0131(200103)44:3<731::AID-ANR124>3.0.CO;2-U.CrossRefPubMed Kubo M, Ihn H, Yamane K, Tamaki K: Up-regulated expression of transforming growth factor β receptors in dermal fibroblasts in skin sections from patients with localized scleroderma. Arthritis Rheum. 2001, 44: 731-734. 10.1002/1529-0131(200103)44:3<731::AID-ANR124>3.0.CO;2-U.CrossRefPubMed
51.
Zurück zum Zitat Pendergrass SA, Hayes E, Farina G, Lemaire R, Farber HW, Whitfield ML, Lafyatis R: Limited systemic sclerosis patients with pulmonary arterial hypertension show biomarkers of inflammation and vascular injury. PLoS ONE. 2010, 5: Pendergrass SA, Hayes E, Farina G, Lemaire R, Farber HW, Whitfield ML, Lafyatis R: Limited systemic sclerosis patients with pulmonary arterial hypertension show biomarkers of inflammation and vascular injury. PLoS ONE. 2010, 5:
52.
Zurück zum Zitat Sato T, Murakumo Y, Hagiwara S, Jijiwa M, Suzuki C, Yatabe Y, Takahashi M: High-level expression of CD109 is frequently detected in lung squamous cell carcinomas. Pathol Int. 2007, 57: 719-724. 10.1111/j.1440-1827.2007.02168.x.CrossRefPubMed Sato T, Murakumo Y, Hagiwara S, Jijiwa M, Suzuki C, Yatabe Y, Takahashi M: High-level expression of CD109 is frequently detected in lung squamous cell carcinomas. Pathol Int. 2007, 57: 719-724. 10.1111/j.1440-1827.2007.02168.x.CrossRefPubMed
53.
Zurück zum Zitat Zhang JM, Hashimoto M, Kawai K, Murakumo Y, Sato T, Ichihara M, Nakamura S, Takahashi M: CD109 expression in squamous cell carcinoma of the uterine cervix. Pathol Int. 2005, 55: 165-169. 10.1111/j.1440-1827.2005.01807.x.CrossRefPubMed Zhang JM, Hashimoto M, Kawai K, Murakumo Y, Sato T, Ichihara M, Nakamura S, Takahashi M: CD109 expression in squamous cell carcinoma of the uterine cervix. Pathol Int. 2005, 55: 165-169. 10.1111/j.1440-1827.2005.01807.x.CrossRefPubMed
54.
Zurück zum Zitat Furst DE, Clements PJ, Steen VD, Medsger TA, Masi AT, D'Angelo WA, Lachenbruch PA, Grau RG, Seibold JR: The modified Rodnan skin score is an accurate reflection of skin biopsy thickness in systemic sclerosis. J Rheumatol. 1998, 25: 84-88.PubMed Furst DE, Clements PJ, Steen VD, Medsger TA, Masi AT, D'Angelo WA, Lachenbruch PA, Grau RG, Seibold JR: The modified Rodnan skin score is an accurate reflection of skin biopsy thickness in systemic sclerosis. J Rheumatol. 1998, 25: 84-88.PubMed
Metadaten
Titel
CD109, a TGF-β co-receptor, attenuates extracellular matrix production in scleroderma skin fibroblasts
verfasst von
Xiao-Yong Man
Kenneth W Finnson
Murray Baron
Anie Philip
Publikationsdatum
01.06.2012
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 3/2012
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/ar3877

Weitere Artikel der Ausgabe 3/2012

Arthritis Research & Therapy 3/2012 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.