Your browser doesn't support javascript.
loading
Cyclophilin A contributes to aortopathy induced by postnatal loss of smooth muscle TGFBR1.
Zhou, Guannan; Liao, Mingmei; Wang, Fen; Qi, Xiaoyan; Yang, Pu; Berceli, Scott A; Sharma, Ashish K; Upchurch, Gilbert R; Jiang, Zhihua.
Afiliação
  • Zhou G; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Liao M; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Wang F; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Qi X; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Yang P; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Berceli SA; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Sharma AK; Malcom Randall Veterans Affairs Medical Center, Gainesville, Florida, USA.
  • Upchurch GR; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
  • Jiang Z; Division of Vascular Surgery and Endovascular Therapy, University of Florida College of Medicine, Gainesville, Florida, USA.
FASEB J ; 33(10): 11396-11410, 2019 10.
Article em En | MEDLINE | ID: mdl-31311317
ABSTRACT
Recent recognition that TGF-ß signaling disruption is involved in the development of aortic aneurysms has led to renewed investigations into the role of TGF-ß biology in the aortic wall. We previously found that the type I receptor of TGF-ß (TGFBR2) receptor contributes to formation of ascending aortic aneurysms and dissections (AADs) induced by smooth muscle cell (SMC)-specific, postnatal deletion of Tgfbr1 (Tgfbr1iko). Here, we aimed to decipher the mechanistic signaling pathway underlying the pathogenic effects of TGFBR2 in this context. Gene expression profiling demonstrated that Tgfbr1iko triggers an acute inflammatory response in developing AADs, and Tgfbr1iko SMCs express an inflammatory phenotype in culture. Comparative proteomics profiling and mass spectrometry revealed that Tgfbr1iko SMCs respond to TGF-ß1 stimulation via robust up-regulation of cyclophilin A (CypA). This up-regulation is abrogated by inhibition of TGFBR2 kinase activity, small interfering RNA silencing of Tgfbr2 expression, or inhibition of SMAD3 activation. In mice, Tgfbr1iko rapidly promotes CypA production in SMCs of developing AADs, whereas treatment with a CypA inhibitor attenuates aortic dilation by 56% (P = 0.003) and ameliorates aneurysmal degeneration (P = 0.016). These protective effects are associated with reduced aneurysm-promoting inflammation. Collectively, these results suggest a novel mechanism, wherein loss of type I receptor of TGF-ß triggers promiscuous, proinflammatory TGFBR2 signaling in SMCs, thereby promoting AAD formation.-Zhou, G., Liao, M., Wang, F., Qi, X., Yang, P., Berceli, S. A., Sharma, A. K., Upchurch, G. R., Jr., Jiang, Z. Cyclophilin A contributes to aortopathy induced by postnatal loss of smooth muscle TGFBR1.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Aorta / Doenças da Aorta / Ciclofilina A / Miócitos de Músculo Liso / Receptor do Fator de Crescimento Transformador beta Tipo I / Músculo Liso Vascular Limite: Animals Idioma: En Revista: FASEB J Ano de publicação: 2019 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Aorta / Doenças da Aorta / Ciclofilina A / Miócitos de Músculo Liso / Receptor do Fator de Crescimento Transformador beta Tipo I / Músculo Liso Vascular Limite: Animals Idioma: En Revista: FASEB J Ano de publicação: 2019 Tipo de documento: Article