Your browser doesn't support javascript.
loading
Hepatobiliary organoids differentiated from hiPSCs relieve cholestasis-induced liver fibrosis in nonhuman primates.
Li, Hongmei; Li, Jingyi; Wang, Ting; Sun, Ke; Huang, Guangrui; Cao, Yulin; Wu, Fenfang; Xu, Anlong.
Afiliação
  • Li H; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
  • Li J; Beizhong Jingyuan Biotechnology (Beijing) Limited, Beijing, People's Republic of China.
  • Wang T; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
  • Sun K; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
  • Huang G; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
  • Cao Y; School of Life Science, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
  • Wu F; Beizhong Jingyuan Biotechnology (Beijing) Limited, Beijing, People's Republic of China.
  • Xu A; Beizhong Jingyuan Biotechnology (Beijing) Limited, Beijing, People's Republic of China.
Int J Biol Sci ; 20(4): 1160-1179, 2024.
Article em En | MEDLINE | ID: mdl-38385067
ABSTRACT
There is an urgent need for novel therapies to treat end-stage liver disease due to the shortage of available organs. Although cell transplantation holds considerable promise, its availability is limited due to the low engrafted cell mass and lack of unifying cell transplantation strategies. Here, we optimally established human induced pluripotent stem cell-derived functional hepatobiliary organoids (HBOs) based on our previous research and transplanted them into a monkey model via liver subcapsular and submesenteric transplantation routes to assess their potential clinical application. Our study revealed that HBO transplantation could safely and effectively improve hepatoprotection effects by antiapoptotic and antifibrotic agents. In addition, we also discovered that while multiple HBO transplantation pathways may have a shared effector mechanism, their respective treatment approaches have distinct advantages. Transplantation of HBOs could promote the high expression of CTSV in hepatic sinusoid endothelial cells, which might halt the progression of hepatic sinusoidal capillarization and liver fibrosis. Liver subcapsular transplants had stronger pro-CTSV upregulation than HBO submesenteric transplants, which could be attributed to naturally high CTSV expression in HBOs. Interestingly, both transplantation routes of HBOs were targeted the injured liver and triggered a new pattern of ductular reaction to alleviate the degree of liver fibrosis by surrounding the area with CK19-positive labeled cells. These residing, homing and repairing effects might be related to the high expression of MMP family genes. By promoting a unique pattern of ductular reactions, submesenteric HBO transplantation has a more representative antifibrotic impact than liver subcapsular transplantation. Altogether, our data strongly imply that the treatment of severe liver diseases with liver subcapsular and submesenteric transplantation of HBOs may be clinically effective and safe. These findings provide new insight into HBOs for further experimental and clinical validation.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Colestase / Células-Tronco Pluripotentes Induzidas Limite: Animals / Humans Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Colestase / Células-Tronco Pluripotentes Induzidas Limite: Animals / Humans Idioma: En Ano de publicação: 2024 Tipo de documento: Article