1
|
Li X, Yang W, Weng Y, Zhao Y, Chen H, Chen Y, Qiu J, Jiang B, Li C, Lai Y. Scutellarin Alleviates CCl 4-Induced Liver Fibrosis by Regulating Intestinal Flora and PI3K/AKT Signaling Axis. Int J Mol Sci 2025; 26:2997. [PMID: 40243656 PMCID: PMC11989008 DOI: 10.3390/ijms26072997] [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: 02/18/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Liver fibrosis is a pathological manifestation of chronic liver disease developing to the terminal stage, and there is a lack of effective therapeutic drugs in clinical practice. Scutellarin (SCU) is a flavonoid extracted from Erigeron breviscapus (Vaniot.) Hand.-Mazz., which has significant anti-liver-fibrosis efficacy, but its mode of action remains incompletely understood. A liver fibrosis model was built with male Sprague Dawley rats induced with the disease by CCl4 to evaluate the therapeutic effect of drugs. 16S rRNA sequencing and metabolomics were used to analyze the regulatory effects of SCU on intestinal flora and host metabolism; antibiotics were administered to eliminate gut microbiota and fecal microbiota transplantation (FMT) experiments were used to verify the mechanism. The mechanistic basis underlying SCU's hepatic anti-fibrotic effects was screened by network pharmacology combined with transcriptomics, combined with molecular docking, qPCR, and WB verification. The results showed that SCU may play an anti-liver-fibrosis role by correcting the imbalance of gut flora and regulating the linoleic acid and purine metabolic pathways. In addition, SCU can downregulate the levels of proteins and genes related to the PI3K/AKT axis. In summary, SCU alleviates liver fibrosis by reversing intestinal flora imbalance, regulating the metabolic profile, and inhibiting the PI3K/AKT axis.
Collapse
Affiliation(s)
- Xin Li
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan (Cultivation), Dali University, Dali 671000, China; (X.L.); (B.J.)
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Wanqi Yang
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Ying Weng
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Yingying Zhao
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Haidong Chen
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Yang Chen
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Jishuang Qiu
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Bei Jiang
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan (Cultivation), Dali University, Dali 671000, China; (X.L.); (B.J.)
| | - Chunyan Li
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| | - Yong Lai
- Yunnan Key Laboratory of Screening and Research on Anti-Pathogenic Plant Resources from Western Yunnan (Cultivation), Dali University, Dali 671000, China; (X.L.); (B.J.)
- College of Pharmacy, Dali University, Dali 671000, China; (W.Y.); (Y.W.); (Y.Z.); (H.C.); (Y.C.); (J.Q.)
| |
Collapse
|
2
|
Tong X, Sun Y, Wang Q, Zhao X, Chen W, Zhang M, Ren Y, Zhao X, Wu X, Zhao J, Sun C, Zheng M, Ren H, Yang Z, Ou X, Jia J, You H. Delicate and thin fibrous septa indicate a regression tendency in metabolic dysfunction-associated steatohepatitis patients with advanced fibrosis. Hepatol Int 2025; 19:166-180. [PMID: 39152361 DOI: 10.1007/s12072-024-10719-w] [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: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatohepatitis (MASH)-related fibrosis is reversible. However, the dynamic morphology change in fibrosis regression remains unclear. We aim to explore the morphological characteristics of fibrosis regression in advanced MASH patients. METHODS Clinical and histological data of 79 biopsy-proved MASH patients with advanced fibrosis (F3-F4) were reviewed. The second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) image technology was used to quantitatively identify the R (regressive) septa from P (progressive) septa and PS (perisinusoidal) fibrosis. Non-invasive tests were used to compare the fibrosis level with and without R septa groups. Transcriptomics was used to explore hub genes and the underlying mechanism of the formation of R septa. RESULTS The R septa were different from the P septa and PS fibrosis in detail collagen quantitation identified by SHG/TPEF technology. The R septa were found in MASH fibrosis-regressed patients, which met the definition of the "Beijing classification". Therefore, patients were divided into two groups according to septa morphology: with R septa (n = 10, 12.7%), and without R septa (n = 69, 87.3%). Patients with R septa had lower values in most non-invasive tests, especially for liver stiffness assessed by TE (12.3 vs. 19.4 kPa, p = 0.010) and FAST (FibroScan®-AST) score (0.43 vs. 0.70, p = 0.003). Transcriptomics analysis showed that the expressions of five hub fibrogenic genes, including Col3A1, BGN, Col4A1, THBS2, and Col4A2 in the R septa group, were significantly lower. CONCLUSIONS The R septa can be differentiated from the P septa and PS fibrosis by quantitative assessment of SHG/TPEF, and it represents a tendency of fibrosis regression in MASH patients. TRIAL REGISTRATION NCT03386890, 29/12/2017.
Collapse
Affiliation(s)
- Xiaofei Tong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Yameng Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Qianyi Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Wei Chen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Mengyang Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Yayun Ren
- HistoIndex Pte Ltd, Singapore, Singapore
| | - Xinyu Zhao
- Clinical Epidemiology and EBM Unit, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Xiaoning Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Jingjie Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Chenglin Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Minghua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Hao Ren
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Ou
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, 95 Yong-An Road, Xi-Cheng District, Beijing, 100050, China.
| |
Collapse
|
3
|
Ramachandran P, Brice M, Sutherland EF, Hoy AM, Papachristoforou E, Jia L, Turner F, Kendall TJ, Marwick JA, Carragher NO, Oro D, Feigh M, Leeming DJ, Nielsen MJ, Karsdal MA, Hartmann N, Erickson M, Adorini L, Roth JD, Fallowfield JA. Aberrant basement membrane production by HSCs in MASLD is attenuated by the bile acid analog INT-767. Hepatol Commun 2024; 8:e0574. [PMID: 39585303 PMCID: PMC11596521 DOI: 10.1097/hc9.0000000000000574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/07/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND The farnesoid X receptor (FXR) is a leading therapeutic target for metabolic dysfunction-associated steatohepatitis (MASH)-related fibrosis. INT-767, a potent FXR agonist, has shown promise in preclinical models. We aimed to define the mechanisms of INT-767 activity in experimental MASH and dissect cellular and molecular targets of FXR agonism in human disease. METHODS Leptin-deficient ob/ob mice were fed a MASH-inducing diet for 15 weeks before the study started. After baseline liver biopsy and stratification, mice were allocated to INT-767 (10 mg/kg/d) or vehicle treatment for 8 weeks, either alongside an ongoing MASH diet (progression) or following conversion to normal chow (reversal). Effects on extracellular matrix remodeling were analyzed histologically and by RNA-sequencing. Serum fibrosis biomarkers were measured longitudinally. Human liver samples were investigated using bulk and single-cell RNA-sequencing, histology, and cell culture assays. RESULTS INT-767 treatment was antifibrotic during MASH progression but not reversal, attenuating the accumulation of type I collagen and basement membrane proteins (type IV collagen and laminin). Circulating levels of PRO-C4, a type IV collagen formation marker, were reduced by INT-767 treatment and correlated with fibrosis. Expression of basement membrane constituents also correlated with fibrosis severity and adverse clinical outcomes in human MASH. Single-cell RNA-sequencing analysis of mouse and human livers, and immunofluorescence staining colocalized FXR and basement membrane expression to myofibroblasts within the fibrotic niche. Treatment of culture-activated primary human HSCs with INT-767 decreased expression of basement membrane components. CONCLUSIONS These findings highlight the importance of basement membrane remodeling in MASH pathobiology and as a source of circulating biomarkers. Basement membrane deposition by activated HSCs is abrogated by INT-767 treatment and measurement of basement membrane molecules should be included when determining the therapeutic efficacy of FXR agonists.
Collapse
Affiliation(s)
- Prakash Ramachandran
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Madara Brice
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Elena F. Sutherland
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anna M. Hoy
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Eleni Papachristoforou
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Li Jia
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Frances Turner
- Edinburgh Genomics, University of Edinburgh, Edinburgh, UK
| | - Timothy J. Kendall
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - John A. Marwick
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Neil O. Carragher
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | - Mary Erickson
- Intercept Pharmaceuticals Inc., San Diego, California, USA
| | | | | | - Jonathan A. Fallowfield
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Ding J, Liu H, Zhang X, Zhao N, Peng Y, Shi J, Chen J, Chi X, Li L, Zhang M, Liu WY, Zhang L, Ouyang J, Yuan Q, Liao M, Tan Y, Li M, Xu Z, Tang W, Xie C, Li Y, Pan Q, Xu Y, Cai SY, Byrne CD, Targher G, Ouyang X, Zhang L, Jiang Z, Zheng MH, Sun F, Chai J. Integrative multiomic analysis identifies distinct molecular subtypes of NAFLD in a Chinese population. Sci Transl Med 2024; 16:eadh9940. [PMID: 39504356 DOI: 10.1126/scitranslmed.adh9940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/07/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become a common health care burden worldwide. The high heterogeneity of NAFLD remains elusive and impairs outcomes of clinical diagnosis and pharmacotherapy. Several NAFLD classifications have been proposed on the basis of clinical, genetic, alcoholic, or serum metabolic analyses. Yet, accurately predicting the progression of NAFLD to cirrhosis or hepatocellular carcinoma (HCC) in patients remains a challenge. Here, on the basis of a Chinese cohort of patients, we classified NAFLD into three distinct molecular subtypes (NAFLD-mSI, NAFLD-mSII, and NAFLD-mSIII) using integrative multiomics including whole-genome sequencing (WGS), proteomics, phosphoproteomics, lipidomics, and metabolomics across a broad range of liver, blood, and urine specimens. We found that NAFLD-mSI had higher expression of CYP1A2 and CYP3A4, which alleviate hepatic steatosis through mediating free fatty acid/bile acid-mTOR-FXR/PPARα signaling. NAFLD-mSII displayed an elevated risk of liver cirrhosis along with increased hepatic infiltration of M1 and M2 macrophages because of lipid-triggered hepatic CCL2 and CRP production. NAFLD-mSIII exhibited a potential risk for HCC development by increased transcription of CEBPB- and ERCC3-regulated oncogenes because of activation of the EGF-EGFR/CHKA/PI3K-PDK1-AKT cascade. Next, we validated the existence of these three NAFLD molecular subtypes in an external cohort comprising 92 patients with NAFLD across three different Chinese hospitals. These findings may aid in understanding the molecular features underlying NAFLD heterogeneity, thereby facilitating clinical diagnosis and treatment strategies with the aim of preventing the development of liver cirrhosis and HCC.
Collapse
Affiliation(s)
- Jingjing Ding
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Huaizheng Liu
- Department of Emergency, the Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiaoxun Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Nan Zhao
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Peng
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Junping Shi
- Department of Infectious Diseases and Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Jinjun Chen
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xiaoling Chi
- Department of Hepatology, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Ling Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mengni Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liangjun Zhang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiafeng Ouyang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qian Yuan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Department of Pharmacy, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Min Liao
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ya Tan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mingqiao Li
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ziqian Xu
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wan Tang
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuanming Xie
- Institute of Hepatobiliary Surgery, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi Li
- Department of Clinical Laboratory, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Qiong Pan
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Xu
- School of Clinical Medicine and the First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Shi-Ying Cai
- Department of Internal Medicine and Liver Center, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Christopher D Byrne
- Southampton National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton and University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore-Don Calabria Hospital, Negrar di Valpolicella 37024, Italy
| | - Xinshou Ouyang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Liqun Zhang
- Department of Clinical Laboratory, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Zhongyong Jiang
- Department of Medical Laboratory, Cheng du Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu 610213, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou 325000, China
| | - Fengjun Sun
- Department of Pharmacy, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jin Chai
- Department of Gastroenterology, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Institute of Digestive Diseases of PLA, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Cholestatic Liver Diseases Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
- Metabolic Dysfunction-Associated Fatty Liver Disease (MASLD) Medical Research Center, the First Affiliated Hospital (Southwest Hospital), Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
5
|
Yang X, Lin H, Wang M, Huang X, Li K, Xia W, Zhang Y, Wang S, Chen W, Zheng C. Identification of key genes and pathways in duck fatty liver syndrome using gene set enrichment analysis. Poult Sci 2024; 103:104015. [PMID: 39003797 PMCID: PMC11298935 DOI: 10.1016/j.psj.2024.104015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
High-laying ducks are often fed high-energy, nutritious feeds to maintain high productivity, which predisposes them to lipid metabolism disorders and the development of fatty liver syndrome (FLS), which seriously affects production performance and has a substantial economic impact on the poultry industry. Therefore, it is necessary to elucidate the mechanisms underlying the development of fatty liver syndrome. In this study, seven Shan Partridge ducks, each with fatty liver syndrome and normal laying ducks, were selected, and Hematoxylin Eosin staining (HE staining), Masson staining, and transcriptome sequencing were performed on liver tissue. In addition to exploring key genes and pathways using conventional analysis methods, we constructed the first Kyoto Encyclopedia of Genes and Genomes (KEGG) database-based predefined gene set containing 12,764 pathways and 16,836 genes and further performed gene set enrichment analysis (GSEA) on the liver transcriptome data. Finally, key nodes and biological processes were identified via the protein-protein interaction (PPI) network. The results showed that the liver in the FL group exhibited steatosis and fibrosis, and a total of 3,663 genes with upregulated expression versus 2,296 downregulated genes were screened by conventional analysis. GSEA analysis and PPI network analysis revealed that the liver in the FL group exhibited disruption of the mitochondrial electron transport chain, leading to decreased oxidative phosphorylation and the secretion of excessive proinflammatory factors amid the continuous accumulation of lipids. Under continuous chronic inflammation, cell cycle arrest triggers apoptosis, while fibrosis becomes more severe, and procarcinogenic genes are activated, leading to the continuous development and deterioration of the liver. In conclusion, the predefined gene set constructed in this study can be used for GSEA, and the identified hub genes provide useful reference data and a solid foundation for the study of the genetic regulatory mechanism of fatty liver syndrome in ducks.
Collapse
Affiliation(s)
- Xue Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Hao Lin
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China; College of Animal Science, Anhui Science and Technology University, Anhui 233100, P.R. China
| | - Mengpan Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China; College of Animal Science & Veterinary Medicine, Tianjin Agricultural University, Tianjin, 300391, P.R. China
| | - Xuebing Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Kaichao Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Weiguang Xia
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Yanan Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Shuang Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Wei Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China
| | - Chuntian Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, P.R. China.
| |
Collapse
|
6
|
Humphreys DT, Lewis A, Pan‐Castillo B, Berti G, Mein C, Wozniak E, Gordon H, Gadhok R, Minicozzi A, ChinAleong J, Feakins R, Giannoulatou E, James LK, Stagg AJ, Lindsay JO, Silver A. Single cell sequencing data identify distinct B cell and fibroblast populations in stricturing Crohn's disease. J Cell Mol Med 2024; 28:e18344. [PMID: 38685679 PMCID: PMC11058334 DOI: 10.1111/jcmm.18344] [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: 09/27/2023] [Revised: 02/20/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
Single cell RNA sequencing of human full thickness Crohn's disease (CD) small bowel resection specimens was used to identify potential therapeutic targets for stricturing (S) CD. Using an unbiased approach, 16 cell lineages were assigned within 14,539 sequenced cells from patient-matched SCD and non-stricturing (NSCD) preparations. SCD and NSCD contained identical cell types. Amongst immune cells, B cells and plasma cells were selectively increased in SCD samples. B cell subsets suggested formation of tertiary lymphoid tissue in SCD and compared with NSCD there was an increase in IgG, and a decrease in IgA plasma cells, consistent with their potential role in CD fibrosis. Two Lumican-positive fibroblast subtypes were identified and subclassified based on expression of selectively enriched genes as fibroblast clusters (C) 12 and C9. Cells within these clusters expressed the profibrotic genes Decorin (C12) and JUN (C9). C9 cells expressed ACTA2; ECM genes COL4A1, COL4A2, COL15A1, COL6A3, COL18A1 and ADAMDEC1; LAMB1 and GREM1. GO and KEGG Biological terms showed extracellular matrix and stricture organization associated with C12 and C9, and regulation of WNT pathway genes with C9. Trajectory and differential gene analysis of C12 and C9 identified four sub-clusters. Intra sub-cluster gene analysis detected 13 co-regulated gene modules that aligned along predicted pseudotime trajectories. CXCL14 and ADAMDEC1 were key markers in module 1. Our findings support further investigation of fibroblast heterogeneity and interactions with local and circulating immune cells at earlier time points in fibrosis progression. Breaking these interactions by targeting one or other population may improve therapeutic management for SCD.
Collapse
Affiliation(s)
- David T. Humphreys
- Victor Chang Cardiac Research InstituteSydneyNew South WalesAustralia
- St Vincent's Clinical SchoolUniversity of New South WalesSydneyNew South WalesAustralia
| | - Amy Lewis
- Centre for Genomics and Child Health, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Belen Pan‐Castillo
- Centre for Genomics and Child Health, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Giulio Berti
- Centre for Genomics and Child Health, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Charles Mein
- Genome Centre, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Eva Wozniak
- Genome Centre, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Hannah Gordon
- Centre for Immunobiology, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Radha Gadhok
- Centre for Immunobiology, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Annamaria Minicozzi
- Department of Colorectal Surgery, Division of Surgery and Perioperative CareThe Royal London HospitalLondonUK
| | | | - Roger Feakins
- Department of Cellular PathologyRoyal Free London NHS Foundation TrustLondonUK
| | - Eleni Giannoulatou
- Victor Chang Cardiac Research InstituteSydneyNew South WalesAustralia
- St Vincent's Clinical SchoolUniversity of New South WalesSydneyNew South WalesAustralia
| | - Louisa K. James
- Centre for Immunobiology, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Andrew J. Stagg
- Centre for Immunobiology, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - James Oliver Lindsay
- Centre for Immunobiology, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| | - Andrew Silver
- Centre for Genomics and Child Health, Blizard InstituteBarts and The London School of Medicine and DentistryLondonUK
| |
Collapse
|
7
|
Mak JHC, Lui DTW, Fong CHY, Cheung CYY, Wong Y, Lee ACH, Hoo RLC, Xu A, Tan KCB, Lam KSL, Lee CH. Serum thrombospondin-2 level changes with liver stiffness improvement in patients with type 2 diabetes. Clin Endocrinol (Oxf) 2024; 100:230-237. [PMID: 38127469 DOI: 10.1111/cen.15010] [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: 10/03/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Baseline circulating thrombospondin-2 (TSP2) level was identified as a potential novel hepatic fibrosis biomarker that associates with development and progression of hepatic fibrosis in patients with nonalcoholic fatty liver disease and type 2 diabetes. Here, we investigated whether circulating TSP2 levels changed with improvement in liver stiffness (LS), which reflects liver fibrosis on transient elastography. DESIGN Serum TSP2 levels were measured in participants from a randomized, open-label intervention study, at baseline and after 24-weeks treatment of either dapagliflozin 10 mg (N = 30) or sitagliptin 100 mg daily (N = 30). Vibration-controlled transient elastography was performed to evaluate the severity of hepatic fibrosis and steatosis using LS and controlled attenuation parameter (CAP), respectively. PATIENTS AND MEASUREMENTS Among all 60 participants with similar clinical characteristics at baseline (mean HbA1c 8.9%, CAP 289 dB/m and LS 5.8 kPa), despite similar HbA1c lowering, treatment with dapagliflozin, but not sitagliptin, led to significant improvements in body weight (BW) (p = .012), CAP (p = .015) and LS (p = .011) after 24 weeks. RESULTS Serum TSP2 level decreased significantly from baseline in dapagliflozin-treated participants (p = .035), whereas no significant change was observed with sitagliptin. In correlation analysis, change in serum TSP2 levels only positively correlated with change in LS (r = .487, p = .006), but not with changes in BW, CAP or HbA1c after dapagliflozin treatment. CONCLUSIONS Serum TSP2 level decreased with LS after dapagliflozin treatment, and was independent of improvements in BW, glycemic control and hepatic steatosis, further supporting the potential of serum TSP2 level as a novel hepatic fibrosis biomarker in type 2 diabetes.
Collapse
Affiliation(s)
- Jimmy Ho Cheung Mak
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David Tak-Wai Lui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Carol Ho-Yi Fong
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chloe Yu-Yan Cheung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ying Wong
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Alan Chun-Hong Lee
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ruby Lai-Chong Hoo
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kathryn Choon-Beng Tan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Karen Siu-Ling Lam
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chi-Ho Lee
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
8
|
Zhang N, Wu X, Zhang W, Sun Y, Yan X, Xu A, Han Q, Yang A, You H, Chen W. Targeting thrombospondin-2 retards liver fibrosis by inhibiting TLR4-FAK/TGF-β signaling. JHEP Rep 2024; 6:101014. [PMID: 38379585 PMCID: PMC10877131 DOI: 10.1016/j.jhepr.2024.101014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 12/13/2023] [Accepted: 01/08/2024] [Indexed: 02/22/2024] Open
Abstract
Background & Aims Thrombospondin-2 (THBS2) expression is associated with liver fibrosis regardless of etiology. However, the role of THBS2 in the pathogenesis of liver fibrosis has yet to be elucidated. Methods The in vivo effects of silencing Thbs2 in hepatic stellate cells (HSCs) were examined using an adeno-associated virus vector (serotype 6, AAV6) containing short-hairpin RNAs targeting Thbs2, under the regulatory control of cytomegalovirus, U6 or the α-smooth muscle promoter, in mouse models of carbon tetrachloride or methionine-choline deficient (MCD) diet-induced liver fibrosis. Crosstalk between THBS2 and toll-like receptor 4 (TLR4), as well as the cascaded signaling, was systematically investigated using mouse models, primary HSCs, and human HSC cell lines. Results THBS2 was predominantly expressed in activated HSCs and dynamically increased with liver fibrosis progression and decreased with regression. Selective interference of Thbs2 in HSCs retarded intrahepatic inflammatory infiltration, steatosis accumulation, and fibrosis progression following carbon tetrachloride challenge or in a dietary model of metabolic dysfunction-associated steatohepatitis. Mechanically, extracellular THBS2, as a dimer, specifically recognized and directly bound to TLR4, activating HSCs by stimulating downstream profibrotic focal adhesion kinase (FAK)/transforming growth factor beta (TGF-β) pathways. Disruption of the THBS2-TLR4-FAK/TGF-β signaling axis notably alleviated HSC activation and liver fibrosis aggravation. Conclusions THBS2 plays a crucial role in HSC activation and liver fibrosis progression through TLR4-FAK/TGF-β signaling in an autocrine manner, representing an attractive potential therapeutic target for liver fibrosis. Impact and implications Thrombospondin-2 (THBS2) is emerging as a factor closely associated with liver fibrosis regardless of etiology. However, the mechanisms by which THBS2 is involved in liver fibrosis remain unclear. Here, we showed that THBS2 plays a prominent role in the pathogenesis of liver fibrosis by activating the TLR4-TGF-β/FAK signaling axis and hepatic stellate cells in an autocrine manner, providing a potential therapeutic target for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ning Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Xiaoning Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Wen Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Yameng Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Xuzhen Yan
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Anjian Xu
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Qi Han
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Aiting Yang
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Wei Chen
- State Key Lab of Digestive Health, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- National Clinical Research Center of Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
9
|
Linssen EC, Demmers J, van Dijk CGM, van Dam R, Nicese MN, Cheng C, de Kort LMO, de Graaf P. Extracellular matrix analysis of fibrosis: A step towards tissue engineering for urethral stricture disease. PLoS One 2023; 18:e0294955. [PMID: 38032942 PMCID: PMC10688748 DOI: 10.1371/journal.pone.0294955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023] Open
Abstract
The urogenital tract is a target for many congenital and acquired diseases, both benign and oncogenic. In males, the urethra that transports urine and semen can be obstructed by a fibrotic disease called urethral stricture disease (USD). In severe USD, the whole organ including the vascular embedding, the corpus spongiosum (CS), is affected. Recurrent or severe USD is treated by reconstructive surgery. Tissue engineering may improve the outcome of urethral reconstruction in patients with complicated USD. Currently in urethral reconstruction only the epithelial layer is replaced, no substitution for the CS is provided, while the CS is important for mechanical support and vascularization. To develop a tissue engineering strategy for the CS, it is necessary to know the protein composition of the CS. As the extracellular matrix (ECM) plays an important role in the formation of fibrosis, we analyzed the distribution and localization of ECM components in human healthy and fibrotic CS tissue using immunohistology. The morphology of components of the elastic network were affected in USD. After decellularization a clear enrichment of proteins belonging to the ECM was found. In the proteomic analysis collagens COL15A1 and COL4A2 as well as inter-alpha-trypsin inhibitor ITIH4 were upregulated in fibrotic samples. The glycoproteins Periostin (POSTN), Microfibrillar-associated protein 5 (MFAP5) and EMILIN2 are downregulated in fibrotic tissue. To our knowledge this is the first proteomic study of ECM proteins of the CS in healthy and in USD. With these results a regenerating approach for tissue engineered CS can be developed, including relevant ECM proteins that reduce fibrosis and promote healthy healing in urethral reconstructive surgery.
Collapse
Affiliation(s)
- Emma C. Linssen
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jeroen Demmers
- Department of Proteomics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Roos van Dam
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maria Novella Nicese
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Cheng
- Department of Nephrology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Petra de Graaf
- Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
10
|
Zhang W, Li YJ, Zhang N, Chen SY, Tong XF, Wang BQ, Huang T, You H, Chen W. Fibroblast-specific adipocyte enhancer binding protein 1 is a potential pathological trigger and prognostic marker for liver fibrosis independent of etiology. J Dig Dis 2023; 24:550-561. [PMID: 37776122 DOI: 10.1111/1751-2980.13230] [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: 05/09/2023] [Revised: 08/30/2023] [Accepted: 09/28/2023] [Indexed: 10/01/2023]
Abstract
OBJECTIVES Aortic carboxypeptidase-like protein (ACLP) is an extracellular protein involved in adipogenesis, epithelial-mesenchymal transition, epithelial cell hyperplasia, and collagen fibrogenesis. This study mainly aimed to analyze the potential role of adipocyte enhancer binding protein 1 (AEBP1), the ACLP-encoding gene, as a pathological target or prognostic marker for liver fibrosis regardless of etiology. METHODS Dysregulation pattern, clinical relevance, and biological significance of AEBP1 gene in liver fibrosis were analyzed using publicly available transcriptomic profiles, different liver fibrosis mouse models, biological databases, and AEBP1 gene silencing followed by RNA sequencing in human hepatic stellate cells (HSCs). RESULTS AEBP1 gene expression was upregulated and positively correlated with liver fibrogenesis independent of etiology, the protein of which was further verified in liver fibrosis mouse models induced by different pathogenic factors. A higher expression of liver AEBP1 gene had the potential to predict poor prognosis in liver fibrosis. Systematic bioinformatic analyses revealed that AEBP1 expression was HSCs-specific and associated with extracellular matrix (ECM) remodeling and its downstream mechanical-chemical signaling transition. AEBP1 knockdown by specific small interfering RNAs (siRNAs) in HSCs inhibited ECM-receptor interaction and immune-related pathways as well as HSC proliferation or activation. CONCLUSION A high expression of AEBP1 was specifically associated with liver fibrosis and was related to a poor prognosis and predicted the role of AEBP1 in HSCs, providing a new insight for understanding AEBP1 in liver fibrosis.
Collapse
Affiliation(s)
- Wen Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Yu Jia Li
- Emory National primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Ning Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Shu Yan Chen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Xiao Fei Tong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Bing Qiong Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Tao Huang
- Beijing Clinical Research Institute, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Wei Chen
- Beijing Clinical Research Institute, Beijing, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Guo Y, Yuan Z, Hu Z, Gao Y, Guo H, Zhu H, Hong K, Cen K, Mai Y, Bai Y, Yang X. Diagnostic model constructed by five EMT-related genes for renal fibrosis and reflecting the condition of immune-related cells. Front Immunol 2023; 14:1161436. [PMID: 37266443 PMCID: PMC10229861 DOI: 10.3389/fimmu.2023.1161436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Background Renal fibrosis is a physiological and pathological characteristic of chronic kidney disease (CKD) to end-stage renal disease. Since renal biopsy is the gold standard for evaluating renal fibrosis, there is an urgent need for additional non-invasive diagnostic biomarkers. Methods We used R package "limma" to screen out differently expressed genes (DEGs) based on Epithelial-mesenchymal transformation (EMT), and carried out the protein interaction network and GO, KEGG enrichment analysis of DEGs. Secondly, the least absolute shrinkage and selection operator (LASSO), random forest tree (RF), and support vector machine-recursive feature elimination (SVM-RFE) algorithms were used to identify candidate diagnostic genes. ROC curves were plotted to evaluate the clinical diagnostic value of these genes. In addition, mRNA expression levels of candidate diagnostic genes were analyzed in control samples and renal fibrosis samples. CIBERSORT algorithm was used to evaluate immune cells level. Additionally, gene set enrichment analysis (GSEA) and drug sensitivity were conducted. Results After obtaining a total of 24 DEGs, we discovered that they were mostly involved in several immunological and inflammatory pathways, including NF-KappaB signaling, AGE-RAGE signaling, and TNF signaling. Five genes (COL4A2, CXCL1, TIMP1, VCAM1, and VEGFA) were subsequently identified as biomarkers for renal fibrosis through machine learning, and their expression levels were confirmed by validation cohort data sets and in vitro RT-qPCR experiment. The AUC values of these five genes demonstrated significant clinical diagnostic value in both the training and validation sets. After that, CIBERSORT analysis showed that these biomarkers were strongly associated with immune cell content in renal fibrosis patients. GSEA also identifies the potential roles of these diagnostic genes. Additionally, diagnostic candidate genes were found to be closely related to drug sensitivity. Finally, a nomogram for diagnosing renal fibrosis was developed. Conclusion COL4A2, CXCL1, TIMP1, VCAM1, and VEGFA are promising diagnostic biomarkers of tissue and serum for renal fibrosis.
Collapse
Affiliation(s)
- Yangyang Guo
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Ziwei Yuan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zujian Hu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuanyuan Gao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hangcheng Guo
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hengyue Zhu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Hong
- Department of General Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Kenan Cen
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Yifeng Mai
- The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuejia Yang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Chen K, Guo W, Li R, Han Y, Gao Q, Wang S. Demethylzeylasteral attenuates hepatic stellate cell activation and liver fibrosis by inhibiting AGAP2 mediated signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154349. [PMID: 35905567 DOI: 10.1016/j.phymed.2022.154349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Liver fibrosis is a common cause of chronic liver disease. If left untreated, it can ultimately develop into liver cirrhosis or hepatocellular carcinoma. However, a direct antifibrotic therapy is currently unavailable. A re-examination of existing chemicals might be a potential strategy for finding more lead compounds against liver fibrosis. Demethylzeylasteral (T-96), a naturally occurring bioactive compound found in Tripterygium wilfordii Hook. f. (TwHf) possesses multiple pharmacological properties. However, its antifibrotic potential has not yet been fully evaluated. PURPOSE This study aimed to investigate the antifibrotic properties of T-96 and its underlying molecular mechanisms. METHODS The antifibrotic properties of T-96 were investigated in three types of hepatic stellate cells (HSCs) and in a CCl4-induced liver fibrosis mouse model. The effect of T-96 on the proliferation, migration, and activation of HSCs was detected using CCK-8 and scratch/wound healing assays. Hepatic inflammation and fibrosis were evaluated by H&E, Masson's trichrome stain, and Sirius Red staining. The expression of inflammatory and fibrogenic genes was detected by quantitative real-time PCR (qRT-PCR) and western blotting. RNA sequencing (RNA-seq) was performed to explore the potential molecular mechanisms mediating the antifibrotic effect of T-96, which was verified by dual-luciferase reporter assay, qRT-PCR, western blotting, immunofluorescence, and immunoprecipitation analysis. RESULTS The T-96 treatment significantly suppressed the proliferation, migration, and activation of HSCs in vitro. The administration of T-96 attenuated hepatic injury, inflammation, and fibrosis progression in mice with CCl4-induced liver fibrosis. In addition, the RNA-seq of fibrotic liver tissues and subsequent functional verification indicated that the key mechanisms of the antifibrotic effect of T-96 were mediated by suppressing the expression of AGAP2 (Arf GAP with GTPase-like domain, ankyrin repeat and PH domain 2), inhibiting the subsequent phosphorylation of focal adhesion kinase (FAK) and protein kinase B (AKT), and finally reducing the expression of fibrosis-related genes. CONCLUSION Our results provide the first insight that T-96 exerts potent antifibrotic effects both in vitro and in vivo by inhibiting the AGAP2 mediated FAK/AKT signaling axis, and that T-96 may serve as a potential therapeutic candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Ke Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Weiran Guo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Rongxin Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Yueqing Han
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Qi Gao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Shuzhen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
13
|
Wu X, Cheung CKY, Ye D, Chakrabarti S, Mahajan H, Yan S, Song E, Yang W, Lee CH, Lam KSL, Wang C, Xu A. Serum Thrombospondin-2 Levels Are Closely Associated With the Severity of Metabolic Syndrome and Metabolic Associated Fatty Liver Disease. J Clin Endocrinol Metab 2022; 107:e3230-e3240. [PMID: 35532410 DOI: 10.1210/clinem/dgac292] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Metabolic associated fatty liver disease (MAFLD) is the hepatic manifestation of obesity-related metabolic syndrome (MetS). Noninvasive biomarkers for monitoring the progression and severity of these metabolic comorbidities are needed. OBJECTIVES To investigate the associations of serum thrombospondin-2 (TSP2) with MetS and MAFLD severity, and the potential diagnostic value of serum TSP2 for identifying at-risk metabolic associated steatohepatitis (MASH). METHODS Blood samples, clinical data, and liver biopsies were collected from consecutively recruited 252 individuals with morbid obesity receiving bariatric surgery. Histopathology samples of liver biopsies were examined in a blinded fashion by 3 independent pathologists. Serum TSP2 levels were measured by enzyme-linked immunosorbent assay. RESULTS Serum TSP2 levels were significantly elevated in MetS (1.58 [1.07-2.20] ng/mL) compared with non-MetS (1.28 [0.84-1.73] ng/mL; P = .006) in obese patients and positively correlated with increasing number of the MetS components, fasting glucose, glycated hemoglobin, fasting insulin, C-peptide, and homeostatic model assessment of insulin resistance after adjustment of conventional confounders. Serum TSP2 levels differentiated MASH (1.74 [1.32-3.09] ng/mL) from the other non-MASH less severe groups: normal liver (1.41 [1.04-1.63] ng/mL), simple steatosis (1.45 [0.89-1.92] ng/mL), and borderline MASH (1.30 [0.99-2.17] ng/mL) (P < .05). Elevated serum TSP2 was positively associated with the severity of hepatic steatosis, inflammation, fibrosis, and abnormal liver function independent of age, sex and adiposity. Furthermore, high serum TSP2 identified at-risk MASH with area under the operating curve of 0.84 (95% CI 0.70-0.98). CONCLUSION Serum TSP2 is closely associated with severity and progression of MetS and MAFLD, and is a promising noninvasive biomarker for differentiating MASH from benign steatosis and identifying at-risk MASH patients among individuals with obesity.
Collapse
Affiliation(s)
- Xuerui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
| | - Cynthia Kwan Yui Cheung
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dewei Ye
- Key Laboratory of Glucolipid Metabolic Diseases of the Ministry of Education, Guangdong Pharmaceutical University, Guangzhou, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, Canada
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, Canada
| | - Hema Mahajan
- Insititue of Clinical Pathology and Medical Research, Pathology West, NSW Health Pathology, Sydney, NSW 2145, Australia
- University of Sydney, New South Wales, Australia
- Western Sydney University, New South Wales, Australia
| | - Sen Yan
- Dr. Everett Chalmers Hospital, New Brunswick, Canada
| | - Erfei Song
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wah Yang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chi Ho Lee
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Karen Siu Ling Lam
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Cunchuan Wang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
14
|
Yu S, Ericson M, Fanjul A, Erion DM, Paraskevopoulou M, Smith EN, Cole B, Feaver R, Holub C, Gavva N, Horman SR, Huang J. Genome-wide CRISPR Screening to Identify Drivers of TGF-β-Induced Liver Fibrosis in Human Hepatic Stellate Cells. ACS Chem Biol 2022; 17:918-929. [PMID: 35274923 PMCID: PMC9016707 DOI: 10.1021/acschembio.2c00006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Liver fibrosis progression in chronic liver disease leads to cirrhosis, liver failure, or hepatocellular carcinoma and often ends in liver transplantation. Even with an increased understanding of liver fibrogenesis and many attempts to generate therapeutics specifically targeting fibrosis, there is no approved treatment for liver fibrosis. To further understand and characterize the driving mechanisms of liver fibrosis, we developed a high-throughput genome-wide CRISPR/Cas9 screening platform to identify hepatic stellate cell (HSC)-derived mediators of transforming growth factor (TGF)-β-induced liver fibrosis. The functional genomics phenotypic screening platform described here revealed the novel biology of TGF-β-induced fibrogenesis and potential drug targets for liver fibrosis.
Collapse
Affiliation(s)
- Shan Yu
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Matthew Ericson
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Andrea Fanjul
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Derek M. Erion
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02139, United States
| | - Maria Paraskevopoulou
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts 02139, United States
| | - Erin N. Smith
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Banumathi Cole
- HemoShear Therapeutics, Inc., Charlottesville, Virginia 22902, United States
| | - Ryan Feaver
- HemoShear Therapeutics, Inc., Charlottesville, Virginia 22902, United States
| | - Corine Holub
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Narender Gavva
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Shane R. Horman
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Jie Huang
- Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| |
Collapse
|
15
|
Aga H, Soultoukis G, Stadion M, Garcia-Carrizo F, Jähnert M, Gottmann P, Vogel H, Schulz TJ, Schürmann A. Distinct Adipogenic and Fibrogenic Differentiation Capacities of Mesenchymal Stromal Cells from Pancreas and White Adipose Tissue. Int J Mol Sci 2022; 23:ijms23042108. [PMID: 35216219 PMCID: PMC8876166 DOI: 10.3390/ijms23042108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Pancreatic steatosis associates with β-cell failure and may participate in the development of type-2-diabetes. Our previous studies have shown that diabetes-susceptible mice accumulate more adipocytes in the pancreas than diabetes-resistant mice. In addition, we have demonstrated that the co-culture of pancreatic islets and adipocytes affect insulin secretion. The aim of this current study was to elucidate if and to what extent pancreas-resident mesenchymal stromal cells (MSCs) with adipogenic progenitor potential differ from the corresponding stromal-type cells of the inguinal white adipose tissue (iWAT). miRNA (miRNome) and mRNA expression (transcriptome) analyses of MSCs isolated by flow cytometry of both tissues revealed 121 differentially expressed miRNAs and 1227 differentially expressed genes (DEGs). Target prediction analysis estimated 510 DEGs to be regulated by 58 differentially expressed miRNAs. Pathway analyses of DEGs and miRNA target genes showed unique transcriptional and miRNA signatures in pancreas (pMSCs) and iWAT MSCs (iwatMSCs), for instance fibrogenic and adipogenic differentiation, respectively. Accordingly, iwatMSCs revealed a higher adipogenic lineage commitment, whereas pMSCs showed an elevated fibrogenesis. As a low degree of adipogenesis was also observed in pMSCs of diabetes-susceptible mice, we conclude that the development of pancreatic steatosis has to be induced by other factors not related to cell-autonomous transcriptomic changes and miRNA-based signals.
Collapse
Affiliation(s)
- Heja Aga
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (H.A.); (M.S.); (M.J.); (P.G.); (H.V.)
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
| | - George Soultoukis
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany;
| | - Mandy Stadion
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (H.A.); (M.S.); (M.J.); (P.G.); (H.V.)
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
| | - Francisco Garcia-Carrizo
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany;
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (H.A.); (M.S.); (M.J.); (P.G.); (H.V.)
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
| | - Pascal Gottmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (H.A.); (M.S.); (M.J.); (P.G.); (H.V.)
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
| | - Heike Vogel
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (H.A.); (M.S.); (M.J.); (P.G.); (H.V.)
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
- Research Group Genetics of Obesity, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany
- Research Group Molecular and Clinical Life Science of Metabolic Diseases, Faculty of Health Sciences Brandenburg, University of Potsdam, 14469 Potsdam, Germany
| | - Tim J. Schulz
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany;
- Institute of Nutritional Sciences, University of Potsdam, 14558 Nuthetal, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558 Nuthetal, Germany; (H.A.); (M.S.); (M.J.); (P.G.); (H.V.)
- German Center for Diabetes Research (DZD), München-Neuherberg, 85764 München, Germany; (G.S.); (T.J.S.)
- Institute of Nutritional Sciences, University of Potsdam, 14558 Nuthetal, Germany
- Correspondence: ; Tel.: +49-33-200-88-2368
| |
Collapse
|
16
|
Excessive hydrogen sulfide causes lung and brain tissue damage by promoting PARP1/Bax and C9 and inhibiting LAMB1. Apoptosis 2022; 27:149-160. [PMID: 35119561 DOI: 10.1007/s10495-021-01705-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 11/02/2022]
Abstract
Excessive hydrogen sulfide (H2S) causes serious damage to human organs and tissues. In this study, we aimed to explore the role and underlying mechanism of excessive H2S in brain and lung tissues. A H2S concentration of 100-800 pm promotes apoptosis and inflammation of brain and lung cells in ICR mice. Mechanistically, a H2S concentration of 100-800 pm upregulates PARP1 and Bax expression in a dose-dependent manner in vivo and in vitro, and functional gain-and-loss experiments verified that an excessive amount of H2S plays a pro-apoptotic role in HT22 and MML1 cells via regulation of PARP1 and Bax in vitro. By combining animal and cell experiments, we clarified that excess H2S promotes the inflammatory response of mouse brain and lung cells by promoting the expression of C9. In addition, the downregulation of LAMB1 by an excessive H2S concentration was confirmed using mass spectrometry and western blotting in vivo and in vitro. Combined with in vitro experiments, we found that an excessive H2S concentration promotes the expression of STAT1 and EGFR in HT22 and MML1 cells by inhibiting the expression of LAMB1. In summary, 100-800 pm H2S causes the brain and lung tissue damage in ICR mice, the underlying mechanisms include H2S induced apoptosis and inflammation of mouse brain and lung cells by upregulation of PARP1/Bax and C9, respectively, and H2S might induce fibrosis of mouse brain and lung cells by downregulation of LAMB1.
Collapse
|
17
|
Wang H, Zheng S, Jiang H, Wang X, Zhou F, Weng Z. Single-cell transcriptomic analysis reveals a novel cell state and switching genes during hepatic stellate cell activation in vitro. J Transl Med 2022; 20:53. [PMID: 35093101 PMCID: PMC8800312 DOI: 10.1186/s12967-022-03263-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/17/2022] [Indexed: 11/10/2022] Open
Abstract
Background The transformation of hepatic stellate cell (HSC) to myofibroblast is a key event during liver fibrogenesis. However, the differentiation trajectory of HSC-to-myofibroblast transition and the switching genes during this process remains not well understood. Methods We applied single-cell sequencing data to reconstruct a single-lineage pseudotime trajectory of HSC transdifferentiation in vitro and analyzed the gene expression patterns along the trajectory. GeneSwitches was used to identify the order of critical gene expression and functional events during HSC activation. Results A novel cell state during HSC activation was revealed and the HSCs belonging to this state may be an important origin of cancer-associated fibroblasts (CAFs). Combining single-cell transcriptomics with GeneSwitches analyses, we identified some distinct switching genes and the order at which these switches take place for the new state of HSC and the classic culture-activated HSC, respectively. Based on the top switching genes, we established a four-gene combination which exhibited highly diagnostic accuracy in predicting advanced liver fibrosis in patients with nonalcoholic fatty liver disease (NAFLD) or hepatitis B (HBV). Conclusion Our study revealed a novel cell state during HSC activation which may be relevant to CAFs, and identified switching genes that may play key roles in HSC transdifferentiation and serve as predictive markers of advanced fibrosis in patients with chronic liver diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03263-4.
Collapse
|
18
|
Expression of Fibrosis-Related Genes in Liver and Kidney Fibrosis in Comparison to Inflammatory Bowel Diseases. Cells 2022; 11:cells11030314. [PMID: 35159124 PMCID: PMC8834113 DOI: 10.3390/cells11030314] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/08/2022] [Accepted: 01/14/2022] [Indexed: 12/21/2022] Open
Abstract
Fibrosis is an important feature of inflammatory bowel diseases (IBD), but its pathogenesis is incompletely understood. Our aim was to identify genes important for fibrosis in IBD by comparison with kidney and liver fibrosis. First, we performed bioinformatics analysis of Gene Expression Omnibus datasets of liver and kidney fibrosis and identified CXCL9, THBS2, MGP, PTPRC, CD52, GZMA, DPT and DCN as potentially important genes with altered expression in fibrosis. We then performed qPCR analysis of the selected genes’ expression on samples of fibrotic kidney, liver, Crohn’s disease (CD) with and without fibrosis and ulcerative colitis (UC), in comparison to corresponding normal tissue. We found significantly altered expression in fibrosis for all selected genes. A significant difference for some genes was observed in CD with fibrosis in comparison to CD without fibrosis and UC. We conclude that similar changes in the expression of selected genes in liver, kidney fibrosis and IBD provide further evidence that fibrosis in IBD might share common mechanisms with other organs, supporting the hypothesis that fibrosis is the common pathway in diseases of various organs. Some genes were already active in IBD with inflammation without fibrosis, suggesting the early activation of profibrotic pathways or overlapping function in fibrosis and inflammation.
Collapse
|
19
|
Chen L, Li Z, Zheng Y, Zhou F, Zhao J, Zhai Q, Zhang Z, Liu T, Chen Y, Qi S. 3D-printed dermis-specific extracellular matrix mitigates scar contraction via inducing early angiogenesis and macrophage M2 polarization. Bioact Mater 2021; 10:236-246. [PMID: 34901542 PMCID: PMC8636711 DOI: 10.1016/j.bioactmat.2021.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/21/2021] [Accepted: 09/04/2021] [Indexed: 12/19/2022] Open
Abstract
Scar contraction frequently happens in patients with deep burn injuries. Hitherto, porcine dermal extracellular matrix (dECM) has supplied microenvironments that assist in wound healing but fail to inhibit scar contraction. To overcome this drawback, we integrate dECM into three-dimensional (3D)-printed dermal analogues (PDA) to prevent scar contraction. We have developed thermally gelled, non-rheologically modified dECM powder (dECMp) inks and successfully transformed them into PDA that was endowed with a micron-scale spatial structure. The optimal crosslinked PDA exhibited desired structure, good mechanical properties as well as excellent biocompatibility. Moreover, in vivo experiments demonstrated that PDA could significantly reduced scar contraction and improved cosmetic upshots of split thickness skin grafts (STSG) than the commercially available dermal templates and STSG along. The PDA has also induced an early, intense neovascularization, and evoked a type-2-like immune response. PDA's superior beneficial effects may attribute to their desired porous structure, the well-balanced physicochemical properties, and the preserved dermis-specific ECM cues, which collectively modulated the expression of genes such as Wnt11, ATF3, and IL1β, and influenced the crucial endogenous signalling pathways. The findings of this study suggest that PDA is a clinical translatable material that possess high potential in reducing scar contraction. Current dermal analogues have supplied microenvironments that assist in wound healing but cannot inhibit scar contraction. dECMp ink was formulated and transformed into PDA endowed with a micron-scale designed spatial structure. The PDAs were neatly superior to split thickness skin grafts and commercial dermal templates in hindering scar contraction. The transcriptome data may reveal how at the molecular level the IS and skin wounds respond to biomaterial stimuli.
Collapse
Affiliation(s)
- Lei Chen
- Department of Burns, Laboratory of General Surgery, The First Affiliated Hospital, SunYat-Sen University, Guangzhou, 510080, China
| | - Zhiyong Li
- School of Materials Science and Engineering, Centre of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, GD Research Centre for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yongtai Zheng
- School of Materials Science and Engineering, Centre of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, GD Research Centre for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou, 510275, China
| | - Fei Zhou
- Department of Burns, Laboratory of General Surgery, The First Affiliated Hospital, SunYat-Sen University, Guangzhou, 510080, China
| | - Jingling Zhao
- Department of Burns, Laboratory of General Surgery, The First Affiliated Hospital, SunYat-Sen University, Guangzhou, 510080, China
| | - Qiyi Zhai
- Department of Burns, Laboratory of General Surgery, The First Affiliated Hospital, SunYat-Sen University, Guangzhou, 510080, China
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, No. 366, South of Jiangnan Boulevard, Guangzhou, 510280, China
| | - Tianrun Liu
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yongming Chen
- School of Materials Science and Engineering, Centre of Functional Biomaterials, Key Laboratory of Polymeric Composite Materials and Functional Materials of Ministry of Education, GD Research Centre for Functional Biomaterials Engineering and Technology, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shaohai Qi
- Department of Burns, Laboratory of General Surgery, The First Affiliated Hospital, SunYat-Sen University, Guangzhou, 510080, China
| |
Collapse
|
20
|
Lefebvre P, Staels B. Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 2021; 17:662-670. [PMID: 34417588 DOI: 10.1038/s41574-021-00538-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The liver is often thought of as a single functional unit, but both its structural and functional architecture make it highly multivalent and adaptable. In any given physiological situation, the liver can maintain metabolic homeostasis, conduct appropriate inflammatory responses, carry out endobiotic and xenobiotic transformation and synthesis reactions, as well as store and release multiple bioactive molecules. Moreover, the liver is a very resilient organ. This resilience means that chronic liver diseases can go unnoticed for decades, yet culminate in life-threatening clinical complications once the adaptive capacity of the liver is overwhelmed. Non-alcoholic fatty liver disease (NAFLD) predisposes individuals to cirrhosis and increases liver-related and cardiovascular disease-related mortality. This Review discusses the accumulating evidence of sexual dimorphism in NAFLD, which is currently rarely considered in preclinical and clinical studies. Increased awareness of the mechanistic causes of hepatic sexual dimorphism could lead to improved understanding of the biological processes that are dysregulated in NAFLD, to the identification of relevant therapeutic targets and to improved risk stratification of patients with NAFLD undergoing therapeutic intervention.
Collapse
Affiliation(s)
- Philippe Lefebvre
- Université Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Bart Staels
- Université Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
21
|
Kozumi K, Kodama T, Murai H, Sakane S, Govaere O, Cockell S, Motooka D, Kakita N, Yamada Y, Kondo Y, Tahata Y, Yamada R, Hikita H, Sakamori R, Kamada Y, Daly AK, Anstee QM, Tatsumi T, Morii E, Takehara T. Transcriptomics Identify Thrombospondin-2 as a Biomarker for NASH and Advanced Liver Fibrosis. Hepatology 2021; 74:2452-2466. [PMID: 34105780 PMCID: PMC8596693 DOI: 10.1002/hep.31995] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS NAFLD is the most common liver disease worldwide. NASH, the progressive form of NAFLD, and advanced fibrosis are associated with poor outcomes. We searched for their noninvasive biomarkers. APPROACH AND RESULTS Global RNA sequencing of liver tissue from 98 patients with biopsy-proven NAFLD was performed. Unsupervised hierarchical clustering well distinguished NASH from nonalcoholic fatty liver (NAFL), and patients with NASH exhibited molecular abnormalities reflecting their pathological features. Transcriptomic analysis identified proteins up-regulated in NASH and/or advanced fibrosis (stage F3-F4), including matricellular glycoprotein thrombospondin-2 (TSP-2), encoded by the thrombospondin 2 (THBS2) gene. The intrahepatic THBS2 expression level showed the highest areas under the receiver operating characteristic curves (AUROCs) of 0.915 and 0.957 for diagnosing NASH and advanced fibrosis, respectively. THBS2 positively correlated with inflammation and ballooning according to NAFLD activity score, serum aspartate aminotransferase and hyaluronic acid (HA) levels, and NAFLD Fibrosis Score (NFS). THBS2 was associated with extracellular matrix and collagen biosynthesis, platelet activation, caspase-mediated cleavage of cytoskeletal proteins, and immune cell infiltration. Serum TSP-2 expression was measured in 213 patients with biopsy-proven NAFLD, was significantly higher in NASH than in NAFL, and increased parallel to fibrosis stage. The AUROCs for predicting NASH and advanced fibrosis were 0.776 and 0.856, respectively, which were comparable to Fibrosis-4 index, serum HA level, and NFS in advanced fibrosis diagnosis. Serum TSP-2 level and platelet count were independent predictors of NASH and advanced fibrosis. Serum TSP-2 levels could stratify patients with NAFLD according to the risk of hepatic complications, including liver cancer and decompensated cirrhotic events. CONCLUSIONS TSP-2 may be a useful biomarker for NASH and advanced fibrosis diagnosis in patients with NAFLD.
Collapse
Affiliation(s)
- Kazuhiro Kozumi
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Takahiro Kodama
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Hiroki Murai
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Sadatsugu Sakane
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Olivier Govaere
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUnited Kingdom
| | - Simon Cockell
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUnited Kingdom
| | - Daisuke Motooka
- Genome Information Research CenterResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Naruyasu Kakita
- Department of Gastroenterology and HepatologyKaizuka City HospitalOsakaJapan
| | - Yukinori Yamada
- Department of Gastroenterology and HepatologyKaizuka City HospitalOsakaJapan
| | - Yasuteru Kondo
- Department of HepatologySendai Kousei HospitalSendaiJapan
| | - Yuki Tahata
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Ryoko Yamada
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Hayato Hikita
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Ryotaro Sakamori
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Ann K. Daly
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUnited Kingdom
| | - Quentin M. Anstee
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUnited Kingdom,Newcastle National Institute for Health Research Biomedical Research CentreNewcastle Upon Tyne Hospitals National Health Service Foundation TrustNewcastle Upon TyneUnited Kingdom
| | - Tomohide Tatsumi
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| | - Eiichi Morii
- Department of PathologyOsaka University Graduate School of MedicineOsakaJapan
| | - Tetsuo Takehara
- Department of Gastroenterology and HepatologyOsaka University Graduate School of MedicineSuitaJapan
| |
Collapse
|
22
|
Lee CH, Seto WK, Lui DTW, Fong CHY, Wan HY, Cheung CYY, Chow WS, Woo YC, Yuen MF, Xu A, Lam KSL. Circulating Thrombospondin-2 as a Novel Fibrosis Biomarker of Nonalcoholic Fatty Liver Disease in Type 2 Diabetes. Diabetes Care 2021; 44:2089-2097. [PMID: 34183428 DOI: 10.2337/dc21-0131] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/24/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Preclinical studies have suggested that thrombospondin-2 (TSP2) is implicated in liver fibrosis. However, the clinical relevance of TSP2 in nonalcoholic fatty liver disease (NAFLD) remains undefined. Here, we investigated the cross-sectional and longitudinal associations of circulating TSP2 levels with advanced fibrosis (F3 or greater [≥FE] fibrosis) in NAFLD. RESEARCH DESIGN AND METHODS Serum TSP2 levels were measured in 820 patients with type 2 diabetes and NAFLD. All participants received vibration-controlled transient elastography (VCTE) at baseline to evaluate their hepatic steatosis and fibrosis using controlled attenuation parameter (CAP) and liver stiffness (LS) measurements, respectively. Among those without advanced fibrosis at baseline, reassessment VCTE was performed to determine whether ≥F3 fibrosis had developed over time. Multivariable logistic regression analysis was used to evaluate the cross-sectional and longitudinal associations of serum TSP2 level with ≥F3 fibrosis. RESULTS Baseline serum TSP2 level was independently associated with the presence of ≥F3 fibrosis (odds ratio [OR] 5.13, P < 0.001). The inclusion of serum TSP2 level significantly improved the identification of ≥F3 fibrosis by clinical risk factors. Over a median follow-up of 1.5 years, 8.8% developed ≥F3 fibrosis. Baseline serum TSP2 level was significantly associated with incident ≥F3 fibrosis (OR 2.82, P = 0.005), independent of other significant clinical risk factors of fibrosis progression, including BMI, platelet count, and CAP at baseline. CONCLUSIONS Circulating TSP2 level was associated with both the presence and the development of advanced fibrosis and might be a potentially useful prognostic biomarker for the development and progression of liver fibrosis in patients with type 2 diabetes and NAFLD.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong.,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong
| | - Wai-Kay Seto
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong.,State Key Laboratory of Liver Research, University of Hong Kong, Hong Kong
| | - David Tak-Wai Lui
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Carol Ho-Yi Fong
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Helen Yilin Wan
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Chloe Yu-Yan Cheung
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Wing-Sun Chow
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Yu-Cho Woo
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Man-Fung Yuen
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong.,State Key Laboratory of Liver Research, University of Hong Kong, Hong Kong
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong .,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong
| | - Karen Siu-Ling Lam
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong .,State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong
| |
Collapse
|
23
|
Yao S, Chen Z, Yu Y, Zhang N, Jiang H, Zhang G, Zhang Z, Zhang B. Current Pharmacological Strategies for Duchenne Muscular Dystrophy. Front Cell Dev Biol 2021; 9:689533. [PMID: 34490244 PMCID: PMC8417245 DOI: 10.3389/fcell.2021.689533] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked neuromuscular disorder caused by the absence of dystrophin protein, which is essential for muscle fiber integrity. Loss of dystrophin protein leads to recurrent myofiber damage, chronic inflammation, progressive fibrosis, and dysfunction of muscle stem cells. There is still no cure for DMD so far and the standard of care is principally limited to symptom relief through glucocorticoids treatments. Current therapeutic strategies could be divided into two lines. Dystrophin-targeted therapeutic strategies that aim at restoring the expression and/or function of dystrophin, including gene-based, cell-based and protein replacement therapies. The other line of therapeutic strategies aims to improve muscle function and quality by targeting the downstream pathological changes, including inflammation, fibrosis, and muscle atrophy. This review introduces the important developments in these two lines of strategies, especially those that have entered the clinical phase and/or have great potential for clinical translation. The rationale and efficacy of each agent in pre-clinical or clinical studies are presented. Furthermore, a meta-analysis of gene profiling in DMD patients has been performed to understand the molecular mechanisms of DMD.
Collapse
Affiliation(s)
- Shanshan Yao
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|