1
|
Fu X, Xiao P, Luo X, Guo N. Transcription factor YY1 accelerates hepatic fibrosis development by activating NLRP3 inflammasome-mediated pyroptosis. Histol Histopathol 2024; 39:1079-1087. [PMID: 38264929 DOI: 10.14670/hh-18-703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Hepatic fibrosis is the basis of multiple liver diseases and may eventually develop into hepatocellular carcinoma. Hepatic stellate cell (HSC) activation is a driving factor of hepatic fibrogenesis. In the liver microenvironment, liver cells and others play a crucial role in HSC activation. The liver tissues of CCl4-induced rats show excessive fibrosis, inflammation, and cell apoptosis. Yin Yang 1 (YY1) was highly expressed in hepatic fibrosis rats and TGF-β1-treated liver cells. In animal experiments, YY1 knockdown effectively attenuated CCl4-induced liver injury and pyroptosis-related IL-1β and IL-18 expression. In cellular experiments, NLRP3 inflammasome-mediated pyroptosis was activated by TGF-β1 treatment, while YY1 knockdown significantly inhibited the activation of the NLRP3 inflammasome, pyroptosis, and the secretion of IL-1β and IL-18. In addition, our data showed that TGF-β1-treated liver cell conditional medium markedly induced HSC activation, which was rescued by YY1 knockdown in liver cells. YY1 overexpression in liver cells contributed to the activation of TGF-β1-treated liver cell conditional medium in HSCs, however, this effect of YY1 was attenuated by NLRP3 inhibition. Overall, YY1 overexpression in liver cells contributed to HSC activation by facilitating IL-1β and IL-18 production via activating NLRP3 inflammasome-mediated pyroptosis, thus aggravating hepatic fibrogenesis. Our data indicate that YY1 may be a novel target for the treatment of hepatic fibrosis and associated liver diseases.
Collapse
Affiliation(s)
- Xiao Fu
- General Medicine Department, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Ping Xiao
- General Medicine Department, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Xin Luo
- General Medicine Department, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China
| | - Ninghong Guo
- Clinical Trial Center, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, PR China.
| |
Collapse
|
2
|
Fu X, Luo X, Xiao P, Guo N. Yin Yang 1 facilitates the activation, inflammation, and extracellular matrix deposition of hepatic stellate cells in hepatic fibrosis. Pathol Int 2024; 74:197-209. [PMID: 38353379 DOI: 10.1111/pin.13410] [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: 08/28/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 04/11/2024]
Abstract
Chronic hepatic diseases often involve fibrosis as a pivotal factor in their progression. This study investigates the regulatory mechanisms of Yin Yang 1 (YY1) in hepatic fibrosis. Our data reveal that YY1 binds to the prolyl hydroxylase domain 1 (PHD1) promoter. Rats treated with carbon tetrachloride (CCl4) display heightened fibrosis in liver tissues, accompanied by increased levels of YY1, PHD1, and the fibrosis marker alpha-smooth muscle actin (α-SMA). Elevated levels of YY1, PHD1, and α-SMA are observed in the liver tissues of CCl4-treated rats, primary hepatic stellate cells (HSCs) isolated from fibrotic liver tissues, and transforming growth factor beta-1 (TGF-β1)-induced HSCs. The human HSC cell line LX-2, upon YY1 overexpression, exhibits enhanced TGF-β1-induced activation, leading to increased expression of extracellular matrix (ECM)-related proteins and inflammatory cytokines. YY1 silencing produces the opposite effect. YY1 exerts a positive regulatory effect on the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway and PHD1 expression. PHD1 silencing rescues the promotion of YY1 in cell activation, ECM-related protein expression, and inflammatory cytokine production in TGF-β1-treated LX-2 cells. Overall, our findings propose a model wherein YY1 facilitates TGF-β1-induced HSC activation, ECM-related protein expression, and inflammatory cytokine production by promoting PHD1 expression and activating the PI3K/AKT signaling pathway. This study positions YY1 as a promising therapeutic target for hepatic fibrosis.
Collapse
Affiliation(s)
- Xiao Fu
- General Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Xin Luo
- General Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Ping Xiao
- General Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Ninghong Guo
- Clinical Trial Center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| |
Collapse
|
3
|
Jujuboside A ameliorates tubulointerstitial fibrosis in diabetic mice through down-regulating the YY1/TGF-β1 signaling pathway. Chin J Nat Med 2022; 20:656-668. [DOI: 10.1016/s1875-5364(22)60200-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Indexed: 11/17/2022]
|
4
|
Ma H, Wang X, Liu X, Wang C, Gao X, Niu J. miR-654-5p Contributes to the Activation and Proliferation of Hepatic Stellate Cells by Targeting RXRα. Front Cell Dev Biol 2022; 10:841248. [PMID: 35465330 PMCID: PMC9019757 DOI: 10.3389/fcell.2022.841248] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
Liver fibrosis (LF) is a major disease that threatens human health. Hepatic stellate cells (HSCs) contribute directly to LF via extracellular matrix (ECM) secretion. Moreover, RXRα is an important nuclear receptor that plays a key regulatory role in HSC activation. Meanwhile, microRNAs (miRNAs) have been identified as significant regulators of LF development. In particular, miR-654-5p is involved in cellular migration and proliferation, and via bioinformatics analysis, has been identified as a potential factor that targets RXRα in humans and in mice. However, the precise relationship between miR-654-5p and RXRα in the context of LF, remains unknown and is the primary focus of the current study. To establish in vitro activated cell model human primary HSCs were cultured in vitro and LX-2 cells were stimulated with recombinant human TGF-β1. mRNA and protein levels of RXRα, miR-654-5p and fibrogenic genes were compared in quiescent and activated HSCs. Moreover, after transfected with miR-654-5p mimics, the expression changes of above related genes in LX-2 cells were estimated. Meanwhile, cell proliferation and apoptosis were detected in miR-654-5p overexpressed LX-2 cells. Simultaneously, the targeted binding between miR-654-5p and RXRα was verified in LX-2 cells. Carbon tetrachloride (CCl4)-induced mouse model with liver fibrosis was use to research the role of the miR-654-5p in vitro. Our results show that miR-654-5p expression levels increased in activated human HSCs and TGFβ-treated LX-2 cells. Moreover, miR-654-5p mimics markedly promoted LX-2 cell proliferation while inhibiting their apoptosis. Accordingly, the expression levels of RXRα are decreased in activated HSCs and LX-2 cells. Additionally, dual-luciferase reporter assay results reveal direct targeting of RXRα by miR-654-5p. Similarly, in vivo miR-654-5p overexpression aggravates LF in mice that are intraperitoneally injected with CCl4. Taken together, our findings elucidated a novel molecular mechanism with potential use for treatment of LF.
Collapse
Affiliation(s)
- Heming Ma
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Xiaomei Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Xu Liu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Chang Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Xiuzhu Gao
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xiuzhu Gao, ; Junqi Niu,
| | - Junqi Niu
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xiuzhu Gao, ; Junqi Niu,
| |
Collapse
|
5
|
Liu H, Qiu Y, Pei X, Chitteti R, Steiner R, Zhang S, Jin ZG. Endothelial specific YY1 deletion restricts tumor angiogenesis and tumor growth. Sci Rep 2020; 10:20493. [PMID: 33235311 PMCID: PMC7686504 DOI: 10.1038/s41598-020-77568-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/22/2020] [Indexed: 12/23/2022] Open
Abstract
Angiogenesis is a physiological process for the formation of new blood vessels from the pre-existing vessels and it has a vital role in the survival and growth of neoplasms. During tumor angiogenesis, the activation of the gene transcriptions in vascular endothelial cells (ECs) plays an essential role in the promotion of EC proliferation, migration, and vascular network development. However, the molecular mechanisms underlying transcriptional regulation of EC and tumor angiogenesis remains to be fully elucidated. Here we report that the transcription factor Yin Yang 1 (YY1) in ECs is critically involved in tumor angiogenesis. First, we utilized a tamoxifen-inducible EC-specific YY1 deficient mouse model and showed that YY1 deletion in ECs inhibited the tumor growth and tumor angiogenesis. Using the in vivo matrigel plug assay, we then found that EC-specific YY1 ablation inhibited growth factor-induced angiogenesis. Furthermore, vascular endothelial growth factor (VEGF)-induced EC migration was diminished in YY1-depleted human umbilical vein endothelial cells (HUVECs). Finally, a rescue experiment revealed that YY1-regulated BMP6 expression in ECs was involved in EC migration. Collectively, our results demonstrate that endothelial YY1 has a crucial role in tumor angiogenesis and suggest that targeting endothelial YY1 could be a potential therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Huan Liu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.,Aab Cardiovascular Research Institute (CVRI), Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
| | - Yikai Qiu
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Ramamurthy Chitteti
- Aab Cardiovascular Research Institute (CVRI), Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
| | - Rebbeca Steiner
- Aab Cardiovascular Research Institute (CVRI), Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA
| | - Shuya Zhang
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China. .,Aab Cardiovascular Research Institute (CVRI), Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA.
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute (CVRI), Department of Medicine, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY, 14642, USA.
| |
Collapse
|
6
|
Zhang S, Liu H, Yin M, Pei X, Hausser A, Ishikawa E, Yamasaki S, Jin ZG. Deletion of Protein Kinase D3 Promotes Liver Fibrosis in Mice. Hepatology 2020; 72:1717-1734. [PMID: 32048304 PMCID: PMC9338785 DOI: 10.1002/hep.31176] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/23/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Liver fibrosis (LF) is a central pathological process that occurs in most types of chronic liver diseases. Advanced LF causes cirrhosis, hepatocellular carcinoma, and liver failure. However, the exact molecular mechanisms underlying the initiation and progression of LF remain largely unknown. APPROACH AND RESULTS This study was designed to investigate the role of protein kinase D3 (PKD3; gene name Prkd3) in the regulation of liver homeostasis. We generated global Prkd3 knockout (Prkd3-/- ) mice and myeloid-cell-specific Prkd3 knockout (Prkd3∆LysM ) mice, and we found that both Prkd3-/- mice and Prkd3∆LysM mice displayed spontaneous LF. PKD3 deficiency also aggravated CCl4 -induced LF. PKD3 is highly expressed in hepatic macrophages (HMs), and PKD3 deficiency skewed macrophage polarization toward a profibrotic phenotype. Activated profibrotic macrophages produced transforming growth factor beta that, in turn, activates hepatic stellate cells to become matrix-producing myofibroblasts. Moreover, PKD3 deficiency decreased the phosphatase activity of SH2-containing protein tyrosine phosphatase-1 (a bona-fide PKD3 substrate), resulting in sustained signal transducer and activator of transcription 6 activation in macrophages. In addition, we observed that PKD3 expression in HMs was down-regulated in cirrhotic human liver tissues. CONCLUSIONS PKD3 deletion in mice drives LF through the profibrotic macrophage activation.
Collapse
Affiliation(s)
- Shuya Zhang
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Huan Liu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Meimei Yin
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Xiuying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Eri Ishikawa
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan,Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Zheng Gen Jin
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
7
|
Xu T, Pan L, Li L, Hu S, Zhou H, Yang C, Yang J, Li H, Liu Y, Meng X, Li J. MicroRNA-708 modulates Hepatic Stellate Cells activation and enhances extracellular matrix accumulation via direct targeting TMEM88. J Cell Mol Med 2020; 24:7127-7140. [PMID: 32463570 PMCID: PMC7339227 DOI: 10.1111/jcmm.15119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/14/2022] Open
Abstract
Transmembrane protein 88 (TMEM88) is a potential 2-transmembrane-type protein that interacts with the PDZ domain of Dishevelled-1 (DVL-1), a crucial component of Wnt signalling pathway through its C-terminal Val-Trp-Val (VWV) motif in Xenopus embryo cells. Since the significant function of β-catenin in liver fibrosis, it is urgent to study the TMEM88 mechanism in liver fibrosis. The current research was for evaluating the function of TMEM88 in the process of the liver fibrosis and clarifying the inherent mechanism. The study found that TMEM88 is decreased in human fibrotic liver tissues. Functionally, TMEM88 significantly reduced the expression levels of α-smooth muscle actin (α-SMA) and collagen type I (Col.I) and repressed extracellular matrix (ECM) accumulation by restoring the balance between matrix metalloproteinases (MMPs) and TIMPs (tissue inhibitor of metalloproteinases). TMEM88 inhibited HSCs proliferation and evaluated the apoptosis of activated LX-2 cells by regulating Wnt3a, Wnt2b and β-catenin of Wnt/β-catenin signalling pathway. Moreover, we demonstrated that miR-708 particularly targeted TMEM88 3'-UTR regions and down-regulated the expression level of TMEM88 in TGF-β1-stimulated LX-2 cells. MiR-708 promoted the generation of ECM and cell activation in activated LX-2 cells. These results determined that miR-708 could promote HSCs activation and enhance ECM accumulation via direct targeting TMEM88 by Wnt/β-catenin signalling pathway. This will provide a potential target for future research in the process of liver fibrosis.
Collapse
Affiliation(s)
- Tao Xu
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Linxin Pan
- The School of Life Science, Anhui Medical University, Hefei, China
| | - Liangyun Li
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Shuang Hu
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Hong Zhou
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China.,Division of Life Sciences and Medicine, Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Chenchen Yang
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China.,Affiliated Psychological Hospital of Anhui Medical University, Anhui Medical University, Hefei, China.,Hefei Fourth People's Hospital, Hefei, China
| | - Junfa Yang
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China.,Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Haodong Li
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yuming Liu
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiaoming Meng
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun Li
- Anhui Provincial laboratory of inflammatory and immunity disease, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China.,The Key Laboratory of Anti-Inflammatory and Immune medicines, Ministry of Education, Hefei, China.,Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Li G, Ma X, Xu L. The roles of zinc finger proteins in non-alcoholic fatty liver disease. LIVER RESEARCH 2020. [DOI: 10.1016/j.livres.2020.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|