[HTML][HTML] Cooperative interaction of CTGF and TGF-β in animal models of fibrotic disease

Q Wang, W Usinger, B Nichols, J Gray, L Xu… - Fibrogenesis & tissue …, 2011 - Springer
Q Wang, W Usinger, B Nichols, J Gray, L Xu, TW Seeley, M Brenner, G Guo, W Zhang…
Fibrogenesis & tissue repair, 2011Springer
Background Connective tissue growth factor (CTGF) is widely thought to promote the
development of fibrosis in collaboration with transforming growth factor (TGF)-β; however,
most of the evidence for its involvement comes from correlative and culture-based studies. In
this study, the importance of CTGF in tissue fibrosis was directly examined in three murine
models of fibrotic disease: a novel model of multiorgan fibrosis induced by repeated
intraperitoneal injections of CTGF and TGF-β2; the unilateral ureteral obstruction (UUO) …
Background
Connective tissue growth factor (CTGF) is widely thought to promote the development of fibrosis in collaboration with transforming growth factor (TGF)-β; however, most of the evidence for its involvement comes from correlative and culture-based studies. In this study, the importance of CTGF in tissue fibrosis was directly examined in three murine models of fibrotic disease: a novel model of multiorgan fibrosis induced by repeated intraperitoneal injections of CTGF and TGF-β2; the unilateral ureteral obstruction (UUO) renal fibrosis model; and an intratracheal bleomycin instillation model of pulmonary fibrosis.
Results
Intraperitoneal coadministration of CTGF and TGF-β2 elicited a profound fibrotic response that was inhibited by the human anti-CTGF antibody FG-3019, as indicated by the ability of FG-3019 to ameliorate the histologic signs of fibrosis and reduce the otherwise increased hydroxyproline:proline (Hyp:Pro) ratios by 25% in kidney (P < 0.05), 30% in liver (P < 0.01) and 63% in lung (P < 0.05). Moreover, administration of either cytokine alone failed to elicit a fibrotic response, thus demonstrating that CTGF is both necessary and sufficient to initiate fibrosis in the presence of TGF-β and vice versa. In keeping with this requirement for CTGF function in fibrosis, FG-3019 also reduced the renal Hyp:Pro response up to 20% after UUO (P < 0.05). In bleomycin-injured animals, a similar trend towards a FG-3019 treatment effect was observed (38% reduction in total lung Hyp, P = 0.056). Thus, FG-3019 antibody treatment consistently reduced excessive collagen deposition and the pathologic severity of fibrosis in all models.
Conclusion
Cooperative interactions between CTGF and TGF-β signaling are required to elicit overt tissue fibrosis. This interdependence and the observed anti-fibrotic effects of FG-3019 indicate that anti-CTGF therapy may provide therapeutic benefit in different forms of fibroproliferative disease.
Springer