1
|
Liu D, Wu Z, Gao J, Mei Q, Zhang X, Wang B. CircUTRN24/miR-483-3p/IGF-1 Regulates Autophagy Mediated Liver Fibrosis in Biliary Atresia. Mol Biotechnol 2024; 66:1424-1433. [PMID: 37369954 DOI: 10.1007/s12033-023-00802-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
Biliary atresia (BA) is a rare neonatal cholestatic disease that presents with a marked bile duct reaction and rapid fibrotic development. Our earlier research has shown that circUTRN24 is highly elevated in BA, but the exact molecular mechanism is still unknown. This study attempted to investigate whether circUTRN24 induces BA liver fibrosis through regulation of autophagy and to elucidate its molecular mechanism. Using TGF-β-treated hepatic stellate cells (HSC) LX-2, we created a liver fibrosis model. qRT-PCR was used to analyze the expression of circUTRN24, miR-483-3p, and IGF-1. Western blot analysis was used to assess the expression of IGF-1, HSC activation-related proteins, and autophagy-related proteins. The TGF-β-induced LX-2 cell fibrosis model was then supplemented with circUTRN24 siRNA, miR-483-3p mimics, and the autophagy activator Rapamycin, and functional rescue tests were carried out to investigate the role of circUTRN24, miR-483-3p, and autophagy in BA liver fibrosis. Using a luciferase reporter assay, a direct interaction between miR-483-3p and circUTRN24 or IGF-1 was discovered. With the increase of TGF-β treatment concentration, circUTRN24 expression also gradually increased, as did HSC activation and autophagy-related protein. si-circUTRN24 significantly decreased circUTRN24 expression and inhibited HSC activation and autophagy, which was reversed by Rapamycin. Through bioinformatics prediction and validation, we found circUTRN24 might act through miR-483-3p targeting IGF-1 in the autophagy-related mTOR pathway. Furthermore, miR-483-3p mimics significantly increased miR-483-3p expression and inhibited HSC activation and autophagy, which were reversed by Rapamycin. Functional rescue experiments showed that si-circUTRN24 inhibited circUTRN24 and IGF-1 expressions and promoted miR-483-3p expression, while the miR-483-3p inhibitor abolished these effects. These findings imply that circUTRN24/miR-483-3p/IGF-1 axis mediated LX-2 cell fibrosis by regulating autophagy.
Collapse
Affiliation(s)
- Dong Liu
- Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Lianhua Street, Futian District, Shenzhen, 518038, Guangdong, China
| | - Zhouguang Wu
- Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Lianhua Street, Futian District, Shenzhen, 518038, Guangdong, China
| | - Jiahui Gao
- Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Lianhua Street, Futian District, Shenzhen, 518038, Guangdong, China
| | - Qianqian Mei
- Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Lianhua Street, Futian District, Shenzhen, 518038, Guangdong, China
| | - Xiyun Zhang
- Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Lianhua Street, Futian District, Shenzhen, 518038, Guangdong, China
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, 7019 Yitian Road, Lianhua Street, Futian District, Shenzhen, 518038, Guangdong, China.
| |
Collapse
|
2
|
Fligor SC, Tsikis ST, Hirsch TI, Jain A, Sun L, Rockowitz S, Gura KM, Puder M. Inflammation drives pathogenesis of early intestinal failure-associated liver disease. Sci Rep 2024; 14:4240. [PMID: 38378873 PMCID: PMC10879484 DOI: 10.1038/s41598-024-54675-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/15/2024] [Indexed: 02/22/2024] Open
Abstract
Patients with intestinal failure who receive long-term parenteral nutrition (PN) often develop intestinal failure-associated liver disease (IFALD). Although there are identified risk factors, the early pathogenesis is poorly understood and treatment options are limited. Here, we perform a transcriptomic analysis of liver tissue in a large animal IFALD model to generate mechanistic insights and identify therapeutic targets. Preterm Yorkshire piglets were provided PN or bottle-fed with sow-milk replacer for 14 days. Compared to bottle-fed controls, piglets receiving PN developed biochemical cholestasis by day of life 15 (total bilirubin 0.2 vs. 2.9 mg/dL, P = 0.01). RNA-Seq of liver tissue was performed. Ingenuity Pathway Analysis identified 747 differentially expressed genes (343 upregulated and 404 downregulated) with an adjusted P < 0.05 and a fold-change of > |1|. Enriched canonical pathways were identified, demonstrating broad activation of inflammatory pathways and inhibition of cell cycle progression. Potential therapeutics including infliximab, glucocorticoids, statins, and obeticholic acid were identified as predicted upstream master regulators that may reverse the PN-induced gene dysregulation. The early driver of IFALD in neonates may be inflammation with an immature liver; identified therapeutics that target the inflammatory response in the liver should be investigated as potential treatments.
Collapse
Affiliation(s)
- Scott C Fligor
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Savas T Tsikis
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Thomas I Hirsch
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Ashish Jain
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA, USA
| | - Liang Sun
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA, USA
| | - Shira Rockowitz
- Harvard Medical School, Boston, MA, USA
- Research Computing, Information Technology, Boston Children's Hospital, Boston, MA, USA
- Division of Genetics and Genomics, and the Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Kathleen M Gura
- Harvard Medical School, Boston, MA, USA
- Department of Pharmacy and the Division of Gastroenterology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Mark Puder
- Vascular Biology Program and Department of Surgery, Boston Children's Hospital, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Liu F, Wei R, Xu X, Lan M, Tao B, Liang Z, Zeng J. Alterations of gut microbiota in infants with biliary atresia identified by 16S rRNA-sequencing. BMC Pediatr 2024; 24:117. [PMID: 38355416 PMCID: PMC10865691 DOI: 10.1186/s12887-024-04582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 01/21/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Biliary atresia (BA) is a severe neonatal disease with progressive intra- and extra-hepatic bile ducts inflammation and hepatic fibrosis. Characterization of gut microbiome profiles in infants with biliary atresia can provide valuable information and potential disease biomarkers. Our study aims to explore the relationship between gut microbiota and biliary atresia. METHODS 16 S ribosomal RNA (rRNA) gene sequencing was carried out to identify the differences in composition and diversity of gut microbiota between infants with BA and healthy subjects. A total of 31 infants with biliary atresia and 20 healthy subjects were recruited. RESULTS The composition of gut microbiota in BA group was significantly different with the normal control group (P < 0.05) and the abundance ratio of Klebsiella/Bifidobacterium showed great potential for identification of BA (P < 0.01). In addition, the differential bacterial taxa were involved in lipid and vitamins metabolism. CONCLUSION Our results could provide potential non-invasive biomarker for identification of biliary atresia and contribute to the treatment in terms of ameliorating microbiota dysbiosis.
Collapse
Affiliation(s)
- Fei Liu
- Department of Pediatric Surgery, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Ru Wei
- Department of Children Health Care, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Xiaogang Xu
- Department of Pediatric Surgery, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Menglong Lan
- Department of Pediatric Surgery, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Boyuan Tao
- Department of Pediatric Surgery, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Zijian Liang
- Department of Pediatric Surgery, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China
| | - Jixiao Zeng
- Department of Pediatric Surgery, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9 Jinsui Road, Guangzhou, Guangdong, 510623, China.
| |
Collapse
|