1
|
Zhang J, Liu G, Liu Y, Yang P, Xie J, Wei X. The biological functions and related signaling pathways of SPON2. Front Oncol 2024; 13:1323744. [PMID: 38264743 PMCID: PMC10803442 DOI: 10.3389/fonc.2023.1323744] [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: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024] Open
Abstract
Spondin-2 (SPON2), also referred to as M-spondin or DIL-1, is a member of the extracellular matrix protein family known as Mindin-F-spondin (FS). SPON2 can be used as a broad-spectrum tumor marker for more than a dozen tumors, mainly prostate cancer. Meanwhile, SPON2 is also a potential biomarker for the diagnosis of certain non-tumor diseases. Additionally, SPON2 plays a pivotal role in regulating tumor metastasis and progression. In normal tissues, SPON2 has a variety of biological functions represented by promoting growth and development and cell proliferation. This paper presents a comprehensive overview of the regulatory mechanisms, diagnostic potential as a broad-spectrum biomarker, diverse biological functions, involvement in various signaling pathways, and clinical applications of SPON2.
Collapse
Affiliation(s)
- Jingrun Zhang
- Zhongshan Clinical College, Dalian University, Dalian, China
- Laboratory of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ge Liu
- Laboratory of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Yuchen Liu
- Zhongshan Clinical College, Dalian University, Dalian, China
| | - Pei Yang
- Department of Neurology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Junyuan Xie
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaowei Wei
- Laboratory of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
2
|
Mindin Activates Autophagy for Lipid Utilization and Facilitates White Spot Syndrome Virus Infection in Shrimp. mBio 2023; 14:e0291922. [PMID: 36779788 PMCID: PMC10127999 DOI: 10.1128/mbio.02919-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023] Open
Abstract
Mindin is a secreted extracellular matrix protein that is involved in regulating cellular events through interacting with integrin. Studies have demonstrated its role in host immunity, including phagocytosis, cell migration, and cytokine production. However, the function of Mindin in the host-virus interaction is largely unknown. In the present study, we report that Mindin facilitates virus infection by activating lipid utilization in an arthropod, kuruma shrimp (Marsupenaeus japonicus). Shrimp Mindin facilitates white spot syndrome virus infection by facilitating viral entry and replication. By activating autophagy, Mindin induces lipid droplet consumption, the hydrolysis of triglycerides into free fatty acids, and ATP production, ultimately providing energy for virus infection. Moreover, integrin is essential for Mindin-mediated autophagy and lipid utilization. Therefore, by revealing the mechanism by which Mindin facilitates virus infection through regulating lipid metabolism, the present study reveals the significance of Mindin in the host-virus interaction. IMPORTANCE White spot syndrome virus (WSSV) is an enveloped double-stranded DNA virus that has had a serious influence on worldwide shrimp farming in the last 30 years. We have demonstrated that WSSV hijacks host autophagy and lipid metabolism for reproduction in kuruma shrimp (Marsupenaeus japonicus). These findings revealed the mechanism by which WSSV exploits host machinery for its infection and provided serial targets for WSSV prevention and control in shrimp farming.
Collapse
|
3
|
Deficiency of mindin reduces renal injury after ischemia reperfusion. Mol Med 2022; 28:152. [PMID: 36510147 PMCID: PMC9743537 DOI: 10.1186/s10020-022-00578-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Acute renal injury (AKI) secondary to ischemia reperfusion (IR) injury continues to be a significant perioperative problem and there is no effective treatment. Mindin belongs to the mindin/F-spondin family and involves in inflammation, proliferation, and cell apoptosis. Previous studies have explored the biological functions of mindin in liver and brain ischemic injury, but its role in AKI is unknown. METHOD To investigate whether mindin has a pathogenic role, mindin knockout (KO) and wild-type (WT) mice were used to establish renal IR model. After 30 min of ischemia and 24 h of reperfusion, renal histology, serum creatinine, and inflammatory response were examined to assess kidney injury. In vitro, proinflammatory factors and inflammatory signaling pathways were measured in mindin overexpression or knockdown and vector cells after hypoxia/reoxygenation (HR). RESULTS Following IR, the kidney mindin level was increased in WT mice and deletion of mindin provided significant protection for mice against IR-induced renal injury as manifested by attenuated the elevation of serum creatinine and blood urea nitrogen along with less severity for histological alterations. Mindin deficiency significantly suppressed inflammatory cell infiltration, TNF-α and MCP-1 production following renal IR injury. Mechanistic studies revealed that mindin deficiency inhibits TLR4/JNK/NF-κB signaling activation. In vitro, the expression levels of TNF-α and MCP-1 were increased in mindin overexpression cells compared with vector cells following HR. Moreover, TLR4/JNK/NF-κB signaling activation was elevated in the mindin overexpression cells in response to HR stimulation while mindin knockdown inhibited the activation of TLR4/JNK/ NF-κB signaling after HR in vitro. Further study showed that mindin protein interacted directly with TLR4 protein. And more, mindin protein was confirmed to be expressed massively in renal tubule tissues of human hydronephrosis patients. CONCLUSION These data demonstrate that mindin is a critical modulator of renal IR injury through regulating inflammatory responses. TLR4/JNK/NF-κB signaling most likely mediates the biological function of mindin in this model of renal ischemia.
Collapse
|
4
|
Non-Alcoholic Fatty Liver Disease (NAFLD) Pathogenesis and Natural Products for Prevention and Treatment. Int J Mol Sci 2022; 23:ijms232415489. [PMID: 36555127 PMCID: PMC9779435 DOI: 10.3390/ijms232415489] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease, affecting approximately one-quarter of the global population, and has become a world public health issue. NAFLD is a clinicopathological syndrome characterized by hepatic steatosis, excluding ethanol and other definite liver damage factors. Recent studies have shown that the development of NAFLD is associated with lipid accumulation, oxidative stress, endoplasmic reticulum stress, and lipotoxicity. A range of natural products have been reported as regulators of NAFLD in vivo and in vitro. This paper reviews the pathogenesis of NAFLD and some natural products that have been shown to have therapeutic effects on NAFLD. Our work shows that natural products can be a potential therapeutic option for NAFLD.
Collapse
|
5
|
Chen Z, Jin ZX, Cai J, Li R, Deng KQ, Ji YX, Lei F, Li HP, Lu Z, Li H. Energy substrate metabolism and oxidative stress in metabolic cardiomyopathy. J Mol Med (Berl) 2022; 100:1721-1739. [PMID: 36396746 DOI: 10.1007/s00109-022-02269-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022]
Abstract
Metabolic cardiomyopathy is an emerging cause of heart failure in patients with obesity, insulin resistance, and diabetes. It is characterized by impaired myocardial metabolic flexibility, intramyocardial triglyceride accumulation, and lipotoxic damage in association with structural and functional alterations of the heart, unrelated to hypertension, coronary artery disease, and other cardiovascular diseases. Oxidative stress plays an important role in the development and progression of metabolic cardiomyopathy. Mitochondria are the most significant sources of reactive oxygen species (ROS) in cardiomyocytes. Disturbances in myocardial substrate metabolism induce mitochondrial adaptation and dysfunction, manifested as a mismatch between mitochondrial fatty acid oxidation and the electron transport chain (ETC) activity, which facilitates ROS production within the ETC components. In addition, non-ETC sources of mitochondrial ROS, such as β-oxidation of fatty acids, may also produce a considerable quantity of ROS in metabolic cardiomyopathy. Augmented ROS production in cardiomyocytes can induce a variety of effects, including the programming of myocardial energy substrate metabolism, modulation of metabolic inflammation, redox modification of ion channels and transporters, and cardiomyocyte apoptosis, ultimately leading to the structural and functional alterations of the heart. Based on the above mechanistic views, the present review summarizes the current understanding of the mechanisms underlying metabolic cardiomyopathy, focusing on the role of oxidative stress.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Zhao-Xia Jin
- Department of Cardiovascular, Huanggang Central Hospital of Yangtze University, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Jingjing Cai
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Central South University, The Third Xiangya Hospital, Changsha, China
| | - Ruyan Li
- Northfield Mount Hermon School, Gill, MA, 01354, USA
| | - Ke-Qiong Deng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Institute of Model Animal, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Fang Lei
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Huo-Ping Li
- Department of Cardiovascular, Huanggang Central Hospital of Yangtze University, Huanggang, China.
- Huanggang Institute of Translational Medicine, Huanggang, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Hongliang Li
- Institute of Model Animal, Wuhan University, Wuhan, China.
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Huanggang Institute of Translational Medicine, Huanggang, China.
- School of Basic Medical Science, Wuhan University, Wuhan, China.
| |
Collapse
|
6
|
Conde de la Rosa L, Goicoechea L, Torres S, Garcia-Ruiz C, Fernandez-Checa JC. Role of Oxidative Stress in Liver Disorders. LIVERS 2022; 2:283-314. [DOI: 10.3390/livers2040023] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Oxygen is vital for life as it is required for many different enzymatic reactions involved in intermediate metabolism and xenobiotic biotransformation. Moreover, oxygen consumption in the electron transport chain of mitochondria is used to drive the synthesis of ATP to meet the energetic demands of cells. However, toxic free radicals are generated as byproducts of molecular oxygen consumption. Oxidative stress ensues not only when the production of reactive oxygen species (ROS) exceeds the endogenous antioxidant defense mechanism of cells, but it can also occur as a consequence of an unbalance between antioxidant strategies. Given the important role of hepatocytes in the biotransformation and metabolism of xenobiotics, ROS production represents a critical event in liver physiology, and increasing evidence suggests that oxidative stress contributes to the development of many liver diseases. The present review, which is part of the special issue “Oxidant stress in Liver Diseases”, aims to provide an overview of the sources and targets of ROS in different liver diseases and highlights the pivotal role of oxidative stress in cell death. In addition, current antioxidant therapies as treatment options for such disorders and their limitations for future trial design are discussed.
Collapse
Affiliation(s)
- Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Leire Goicoechea
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - José C. Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
7
|
Arulkumar R, Jung HJ, Noh SG, Chung HY. Soyasapogenol C from Fermented Soybean ( Glycine Max) Acting as a Novel AMPK/PPARα Dual Activator Ameliorates Hepatic Steatosis: A Novel SANDA Methodology. Int J Mol Sci 2022; 23:5468. [PMID: 35628280 PMCID: PMC9141180 DOI: 10.3390/ijms23105468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Soyasapogenol C (SSC), a derivative of soyasapogenol B (SSB), is specifically found high in many fermented soybean (Glycine max) products, including Cheonggukjang (in Korean). However, the biological activities for preventing and treating hepatic steatosis, and the precise underlying mechanisms of SSC, remain to be explored. (2) Methods: A novel SANDA (structural screening, ADMET prediction, network pharmacology, docking validation, and activity evaluation) methodology was used to examine whether SSC exerts hepatoprotective effects in silico and in vitro. (3) Results: SSC had better ADMET characteristics and a higher binding affinity with predicted targets chosen from network pathway analysis than SSB. SSC induced the phosphorylation of AMP-activated protein kinase (AMPK) and stimulated the nuclear translocation of peroxisome proliferator-activated receptor alpha (PPARα), further enhancing PPAR response element (PPRE) binding activity in HepG2 cells. Concurrently, SSC significantly inhibited triglyceride accumulation, which was associated with the suppression of lipogenesis genes and the enhancement of fatty acid oxidation gene expression in HepG2 cells. (4) Conclusions: Soyasapogenol C, discovered using a novel SANDA methodology from fermented soybean, is a novel AMPK/PPARα dual activator that is effective against hepatic steatosis. Dietary supplementation with soyasapogenol C may prevent the development of hepatic steatosis and other diseases associated with fat accumulation in the liver.
Collapse
Affiliation(s)
- Radha Arulkumar
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, Busan 46241, Korea; (R.A.); (S.G.N.)
| | - Hee Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea;
| | - Sang Gyun Noh
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, Busan 46241, Korea; (R.A.); (S.G.N.)
| | - Hae Young Chung
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, Busan 46241, Korea; (R.A.); (S.G.N.)
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
8
|
Immunity as Cornerstone of Non-Alcoholic Fatty Liver Disease: The Contribution of Oxidative Stress in the Disease Progression. Int J Mol Sci 2021; 22:ijms22010436. [PMID: 33406763 PMCID: PMC7795122 DOI: 10.3390/ijms22010436] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome and has become the major cause of chronic liver disease, especially in western countries. NAFLD encompasses a wide spectrum of hepatic histological alterations, from simple steatosis to steatohepatitis and cirrhosis with a potential development of hepatocellular carcinoma. Non-alcoholic steatohepatitis (NASH) is characterized by lobular inflammation and fibrosis. Several studies reported that insulin resistance, redox unbalance, inflammation, and lipid metabolism dysregulation are involved in NAFLD progression. However, the mechanisms beyond the evolution of simple steatosis to NASH are not clearly understood yet. Recent findings suggest that different oxidized products, such as lipids, cholesterol, aldehydes and other macromolecules could drive the inflammation onset. On the other hand, new evidence indicates innate and adaptive immunity activation as the driving force in establishing liver inflammation and fibrosis. In this review, we discuss how immunity, triggered by oxidative products and promoting in turn oxidative stress in a vicious cycle, fuels NAFLD progression. Furthermore, we explored the emerging importance of immune cell metabolism in determining inflammation, describing the potential application of trained immune discoveries in the NASH pathological context.
Collapse
|
9
|
Bai S, Chaurasiya AH, Banarjee R, Walke PB, Rashid F, Unnikrishnan AG, Kulkarni MJ. CD44, a Predominant Protein in Methylglyoxal-Induced Secretome of Muscle Cells, is Elevated in Diabetic Plasma. ACS OMEGA 2020; 5:25016-25028. [PMID: 33043179 PMCID: PMC7542587 DOI: 10.1021/acsomega.0c01318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/16/2020] [Indexed: 06/11/2023]
Abstract
Methylglyoxal (MG), a glycolytic intermediate and reactive dicarbonyl, is responsible for exacerbation of insulin resistance and diabetic complication. In this study, MG-induced secretome of rat muscle cells was identified and relatively quantified by SWATH-MS. A total of 643 proteins were identified in MG-induced secretome, of which 82 proteins were upregulated and 99 proteins were downregulated by more than 1.3-fold in SWATH analysis. Further, secretory proteins from the classical secretory pathway and nonclassical secretory pathway were identified using SignalP and SecretomeP, respectively. A total of 180 proteins were identified with SignalP, and 113 proteins were identified with SecretomeP. The differentially expressed proteins were functionally annotated by KEGG pathway analysis using Cytoscape software with plugin clusterMaker. The differentially expressed proteins were found to be involved in various pathways like extracellular matrix (ECM)-receptor interaction, leukocyte transendothelial migration, fluid shear stress and atherosclerosis, complement and coagulation cascades, and lysosomal pathway. Since the MG levels are high in diabetic conditions, the presence of MG-induced secreted proteins was inspected by profiling human plasma of healthy and diabetic subjects (n = 10 each). CD44, a predominant MG-induced secreted protein, was found to be elevated in the diabetic plasma and to have a role in the development of insulin resistance.
Collapse
Affiliation(s)
- Shakuntala Bai
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Arvindkumar H. Chaurasiya
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Reema Banarjee
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
| | - Prachi B. Walke
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Faraz Rashid
- Sciex, 121 DHR, Udyog Vihar, Phase IV, Gurugram 122015, Haryana, India
| | | | - Mahesh J. Kulkarni
- Proteomics
Facility, Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| |
Collapse
|
10
|
Inai Y, Ueda K, Matsui ISL, Tajiri M, Minakata S, Wada Y, Ihara Y. Role of C-mannosylation in the secretion of mindin. Biochim Biophys Acta Gen Subj 2020; 1864:129632. [DOI: 10.1016/j.bbagen.2020.129632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022]
|
11
|
Cheng XS, Huo YN, Fan YY, Xiao CX, Ouyang XM, Liang LY, Lin Y, Wu JF, Ren JL, Guleng B. Mindin serves as a tumour suppressor gene during colon cancer progression through MAPK/ERK signalling pathway in mice. J Cell Mol Med 2020; 24:8391-8404. [PMID: 32614521 PMCID: PMC7412704 DOI: 10.1111/jcmm.15332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 01/14/2020] [Accepted: 02/06/2020] [Indexed: 12/13/2022] Open
Abstract
Mindin is important in broad spectrum of immune responses. On the other hand, we previously reported that mindin attenuated human colon cancer development by blocking angiogenesis through Egr-1-mediated regulation. However, the mice original mindin directly suppressed the syngenic colorectal cancer (CRC) growth in our recent study and we aimed to further define the role of mindin during CRC development in mice. We established the mouse syngeneic CRC CMT93 and CT26 WT cell lines with stable mindin knock-down or overexpression. These cells were also subcutaneously injected into C57BL/6 and BALB/c mice as well as established a colitis-associated colorectal cancer (CAC) mouse model treated with lentiviral-based overexpression and knocked-down of mindin. Furthermore, we generated mindin knockout mice using a CRISPR-Cas9 system with CAC model. Our data showed that overexpression of mindin suppressed cell proliferation in both of CMT93 and CT26 WT colon cancer cell lines, while the silencing of mindin promoted in vitro cell proliferation via the ERK and c-Fos pathways and cell cycle control. Moreover, the overexpression of mindin significantly suppressed in vivo tumour growth in both the subcutaneous transplantation and the AOM/DSS-induced CAC models. Consistently, the silencing of mindin reversed these in vivo observations. Expectedly, the tumour growth was promoted in the CAC model on mindin-deficient mice. Thus, mindin plays a direct tumour suppressive function during colon cancer progression and suggesting that mindin might be exploited as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Xiao-Shen Cheng
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Ya-Ni Huo
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Yan-Yun Fan
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Chuan-Xing Xiao
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Xiao-Mei Ouyang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Lai-Ying Liang
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Ying Lin
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Jian-Feng Wu
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Jian-Lin Ren
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China
| | - Bayasi Guleng
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, China.,School of Medicine, Cancer Research Center & Institute of Microbial Ecology, Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| |
Collapse
|
12
|
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med 2020; 152:116-141. [PMID: 32156524 DOI: 10.1016/j.freeradbiomed.2020.02.025] [Citation(s) in RCA: 784] [Impact Index Per Article: 156.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide and is strongly associated with the presence of oxidative stress. Disturbances in lipid metabolism lead to hepatic lipid accumulation, which affects different reactive oxygen species (ROS) generators, including mitochondria, endoplasmic reticulum, and NADPH oxidase. Mitochondrial function adapts to NAFLD mainly through the downregulation of the electron transport chain (ETC) and the preserved or enhanced capacity of mitochondrial fatty acid oxidation, which stimulates ROS overproduction within different ETC components upstream of cytochrome c oxidase. However, non-ETC sources of ROS, in particular, fatty acid β-oxidation, appear to produce more ROS in hepatic metabolic diseases. Endoplasmic reticulum stress and NADPH oxidase alterations are also associated with NAFLD, but the degree of their contribution to oxidative stress in NAFLD remains unclear. Increased ROS generation induces changes in insulin sensitivity and in the expression and activity of key enzymes involved in lipid metabolism. Moreover, the interaction between redox signaling and innate immune signaling forms a complex network that regulates inflammatory responses. Based on the mechanistic view described above, this review summarizes the mechanisms that may account for the excessive production of ROS, the potential mechanistic roles of ROS that drive NAFLD progression, and therapeutic interventions that are related to oxidative stress.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Ruifeng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
13
|
Catalpol Attenuates Hepatic Steatosis by Regulating Lipid Metabolism via AMP-Activated Protein Kinase Activation. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6708061. [PMID: 32420361 PMCID: PMC7201822 DOI: 10.1155/2020/6708061] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/12/2020] [Accepted: 04/04/2020] [Indexed: 12/18/2022]
Abstract
The increased prevalence of nonalcoholic fatty liver disease (NAFLD), which develops from hepatic steatosis, represents a public health challenge. Catalpol, a natural component extracted from the roots of Radix Rehmanniae, has several pharmacological activities. The present study is aimed at examining whether catalpol prevents hepatic steatosis in cell and animal experiments and elucidating the possible mechanisms. HepG2 cells were treated with 300 μM palmitate (PA) and/or catalpol for 24 h in vitro, and male C57BL/6J mice fed a high-fat diet (HFD) were administered catalpol for 18 weeks in vivo. The results revealed that catalpol significantly decreased lipid accumulation in PA-treated HepG2 cells. Moreover, catalpol drastically reduced body weight and lipid accumulation in the liver, whereas it ameliorated hepatocyte steatosis in HFD-fed mice. Notably, catalpol remarkably promoted the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase. Subsequently, catalpol repressed the expressions of lipogenesis-associated genes such as sterol regulatory element-binding protein 1c and fatty acid synthase but promoted the expressions of genes associated with fatty acid β-oxidation such as peroxisome proliferator-activated receptor α together with its target genes carnitine palmitoyltransferase 1 and acyl-CoA oxidase 1 (ACOX1). However, the preincubation of the HepG2 cells with compound C (10 μM), an AMPK inhibitor, prevented catalpol-mediated beneficial effects. These findings suggest that catalpol ameliorates hepatic steatosis by suppressing lipogenesis and enhancing fatty acid β-oxidation in an AMPK-dependent manner. Therefore, catalpol has potential as a novel agent in the treatment of NAFLD.
Collapse
|
14
|
Yang K, Li W, Bai T, Xiao Y, Yu W, Luo P, Cheng F. Mindin deficiency alleviates renal fibrosis through inhibiting NF-κB and TGF-β/Smad pathways. J Cell Mol Med 2020; 24:5740-5750. [PMID: 32253812 PMCID: PMC7214143 DOI: 10.1111/jcmm.15236] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Renal fibrosis acts as a clinical predictor in patients with chronic kidney disease and is characterized by excessive extracellular matrix (ECM) accumulation. Our previous study suggested that mindin can function as a mediator for liver steatosis pathogenesis. However, the role of mindin in renal fibrosis remains obscure. Here, tumour necrosis factor (TGF)‐β‐treated HK‐2 cells and global mindin knockout mouse were induced with renal ischaemia reperfusion injury (IRI) to test the relationship between mindin and renal fibrosis. In vitro, mindin overexpression promoted p65—the hub subunit of the NF‐κB signalling pathway—translocation from the cytoplasm into the nucleus, resulting in NF‐κB pathway activation in TGF‐β‐treated HK‐2 cells. Meanwhile, mindin activated the TGF‐β/Smad pathway, thereby causing fibrotic‐related protein expression in vitro. Mindin−/− mice exhibited less kidney lesions than controls, with small renal tubular expansion, inflammatory cell infiltration, as well as collagen accumulation, following renal IRI. Mechanistically, mindin−/− mice suppressed p65 translocation and deactivated NF‐κB pathway. Simultaneously, mindin disruption inhibited the TGF‐β/Smad pathway, alleviating the expression of ECM‐related proteins. Hence, mindin may be a novel target of renal IRI in the treatment of renal fibrogenesis.
Collapse
Affiliation(s)
- Kang Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Li
- Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Bai
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yusha Xiao
- Department of General Surgery, Research Center of Digestive Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pengcheng Luo
- Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Chen Z, Yu Y, Cai J, Li H. Emerging Molecular Targets for Treatment of Nonalcoholic Fatty Liver Disease. Trends Endocrinol Metab 2019; 30:903-914. [PMID: 31597607 DOI: 10.1016/j.tem.2019.08.006] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
In parallel with the obesity epidemic, nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide. Disequilibrium of lipid metabolism and the subsequent metabolic-stress-induced inflammation are believed to be central in the pathogenesis of NAFLD. Of note, metabolic inflammation is primarily mediated by innate immune signaling, which is increasingly recognized as a driving force in NAFLD progression. Currently, a series of agents targeting one or more of these pathomechanisms have shown encouraging results in preclinical models and clinical trials. This review summarizes the emerging molecular targets involved in signaling in the lipid metabolism and innate immunity aspects of NAFLD, focusing on their mechanistic roles and translational potentials.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China
| | - Yao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China; Basic Medical School, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
16
|
Cai J, Zhang XJ, Li H. The Role of Innate Immune Cells in Nonalcoholic Steatohepatitis. Hepatology 2019; 70:1026-1037. [PMID: 30653691 DOI: 10.1002/hep.30506] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 01/05/2019] [Indexed: 02/06/2023]
Abstract
Inflammation and metabolic dysfunction are hallmarks of nonalcoholic steatohepatitis (NASH), which is one of the fastest-growing liver diseases worldwide. Emerging evidence indicates that innate immune mechanisms are pivotal drivers of inflammation and other pathological manifestations observed in NASH, such as hepatosteatosis, insulin resistance (IR), and fibrosis. This robust innate immune reaction is intrinsic to the liver, which is an important immunological organ that contains a coordinated network of innate immune cells, including Kupffer cells (KCs), dendritic cells (DCs), and lymphocytes. Hepatocytes and liver sinusoidal endothelial cells (LSECs) are not formally innate immune cells, but they take on immune cell function when stressed. These cells can sense excess metabolites and bacterial products and translate those signals into immune responses and pathological hepatic changes during the development of NASH. In this review, we take a historical perspective in describing decades of research that aimed to identify the key molecular and cellular players in the innate immune system in the setting of NASH. Furthermore, we summarize the innate immune cells that are involved in the progression of NASH and illustrate how they sense disturbances in circulating metabolic factors by innate immune receptors and subsequently initiate the intercellular signaling cascades that lead to persistent inflammation and progression of hepatic complications.
Collapse
Affiliation(s)
- Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Wang YY, Shen D, Zhao LJ, Zeng N, Hu TH. Sting is a critical regulator of spinal cord injury by regulating microglial inflammation via interacting with TBK1 in mice. Biochem Biophys Res Commun 2019; 517:741-748. [PMID: 31400857 DOI: 10.1016/j.bbrc.2019.07.125] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological condition that results in progressive tissue loss, secondary to vascular dysfunction and inflammation. Lack of effective pharmacotherapies for SCI is mainly attributable to an incomplete understanding of its pathogenesis. Stimulator of interferon gene (Sting), also known as Transmembrane protein 173 (TMEM173), activates the type I interferon-regulated innate immune response, playing crucial role in modulating inflammation. However, the mechanism underlying Sting activation in SCI is still unclear. Here, we reported that Sting functioned as a positive regulator of SCI. Sting expression was increased in the injured spinal cord samples of SCI mice, along with significantly up-regulated levels of pro-inflammatory cytokines including tumor necrosis factor α (TNF-α), interleukin (IL)-1β and IL-6. Suppressing Sting expression in lipopolysaccharide-incubated mouse microglia markedly reduced the activation of nuclear factor-κB (NF-κB) and mitogen activated protein kinases (MAPKs) signaling pathways, as illustrated by the decreased phosphorylation of IKKβ, IκBα, NF-κB/p65, p38, ERK1/2 and JNK. Furthermore, LPS-stimulated release of pro-inflammatory cytokines in microglial cells was also reversed by Sting knockdown. In contrast, LPS-induced inflammation was further accelerated in microglial cells with Sting over-expression through potentiating NF-κB and MAPKs signaling. Mechanistically, Sting directly interacted with the TANK-binding kinase 1 (TBK1), thus promoting its phosphorylation and the activation of down-streaming NF-κB and MAPKs signaling pathways. Notably, the effects of Sting on SCI progression were verified in mice. Consistently, Sting knockout alleviated inflammatory response and facilitated recovery after SPI in mice through blocking TBK1 activation and subsequent NF-κB and MAPKs phosphorylation. In summary, our findings may provide a novel strategy for prevention and treatment of SCI by targeting Sting.
Collapse
Affiliation(s)
- Yue-Yi Wang
- Second Department of Orthopaed, Ningbo Fenghua District Hospital of Traditional Chinese Medicine, Ningbo, 315040, China.
| | - Dong Shen
- Second Department of Orthopaed, Ningbo Fenghua District Hospital of Traditional Chinese Medicine, Ningbo, 315040, China
| | - Liu-Jun Zhao
- Department of Spine Surgery, Ningbo Sixth Hospital, Ningbo, 315040, China
| | - Nian Zeng
- Second Department of Orthopaed, Ningbo Fenghua District Hospital of Traditional Chinese Medicine, Ningbo, 315040, China
| | - Teng-Hui Hu
- Second Department of Orthopaed, Ningbo Fenghua District Hospital of Traditional Chinese Medicine, Ningbo, 315040, China
| |
Collapse
|
18
|
Liang Z, Xu J, Gu C. Novel role of the SRY-related high-mobility-group box D gene in cancer. Semin Cancer Biol 2019; 67:83-90. [PMID: 31356865 DOI: 10.1016/j.semcancer.2019.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/28/2019] [Accepted: 07/16/2019] [Indexed: 12/18/2022]
Abstract
The SRY-related high-mobility-group box (Sox) gene family encodes a set of transcription factors and is defined by the presence of highly conserved domains. The Sox gene can be divided into 10 groups (A-J). The SoxD subpopulation consists of Sox5, Sox6, Sox13 and Sox23, which are involved in the transcriptional regulation of developmental processes, including embryonic development, nerve growth and cartilage formation. Recently, the SoxD gene family was recognized as important transcriptional regulators associated with many types of cancer. In addition, Sox5 and Sox6 are representatives of the D subfamily, and there are many related studies; however, there are few reports on Sox13 and Sox23. In this review, we first introduce the structures of the SoxD genes. Next, we summarize the latest research progress on SoxD in various types of cancer. Finally, we discuss the potential direction of future SoxD research. In general, the information reviewed here may contribute to future experimental design and increase the potential of SoxD as a cancer treatment target.
Collapse
Affiliation(s)
- Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou 450052, China.
| | - Jing Xu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou 450052, China
| | - Chunhu Gu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
19
|
A K, Uddandrao VVS, Parim B, Ganapathy S, P R N, Kancharla SC, P R, K S, Sasikumar V. Reversal of high fat diet-induced obesity through modulating lipid metabolic enzymes and inflammatory markers expressions in rats. Arch Physiol Biochem 2019; 125:228-234. [PMID: 29553847 DOI: 10.1080/13813455.2018.1452036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
In this study, we evaluated the ameliorative potential of Cucurbita maxima seeds oil (CSO (100 mg/kg body weight)) supplementation to high fat diet (HFD)-induced obese rats for 30 days on the changes in body weight, markers of lipid metabolism such as LDL, HDL, triglycerides, total cholesterol, adiponectin, leptin, amylase, and lipase. We also investigated the effects of CSO on the changes of lipid metabolic enzymes such as fatty-acid synthase, acetyl CoA carboxylase, carnitine palmitoyl transferase-1, HMG CoA reductase, and inflammatory markers (TNF-α and IL-6). Administration of CSO revealed significant diminution in body weight gain, altered the activity, expressions of lipid marker enzymes and inflammatory markers. It demonstrated that CSO had considerably altered these parameters when evaluated with HFD control rats. In conclusion, this study suggested that CSO might ameliorate the HFD-induced obesity by altering the enzymes and mRNA expressions important to lipid metabolism.
Collapse
Affiliation(s)
- Kalaivani A
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
- b Department of Biochemistry , PGP College of Art and Science , Namakkal , India
| | - V V Sathibabu Uddandrao
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
| | - Brahmanaidu Parim
- c ICMR-National Animal Resource Facility for Biomedical Research (NARFBR) , Hyderabad , India
| | - Saravanan Ganapathy
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
| | - Nivedha P R
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
| | | | - Rameshreddy P
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
| | - Swapna K
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
| | - Vadivukkarasi Sasikumar
- a Department of Biochemistry , Centre for Biological Sciences, K. S. Rangasamy College of Arts and Science (Autonomous) , Tiruchengode , India
| |
Collapse
|
20
|
Buffalo liver transcriptome analysis suggests immune tolerance as its key adaptive mechanism during early postpartum negative energy balance. Funct Integr Genomics 2019; 19:759-773. [DOI: 10.1007/s10142-019-00676-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/03/2019] [Accepted: 04/01/2019] [Indexed: 01/25/2023]
|
21
|
Ye P, Liu J, Xu W, Liu D, Ding X, Le S, Zhang H, Chen S, Chen M, Xia J. Dual-Specificity Phosphatase 26 Protects Against Nonalcoholic Fatty Liver Disease in Mice Through Transforming Growth Factor Beta-Activated Kinase 1 Suppression. Hepatology 2019; 69:1946-1964. [PMID: 30582764 PMCID: PMC6594223 DOI: 10.1002/hep.30485] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 12/12/2018] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), which has a wide global distribution, includes different stages ranging from simple steatosis to nonalcoholic steatohepatitis, advanced fibrosis, and liver cirrhosis according to the degree of severity. Chronic low-grade inflammation, insulin resistance, and lipid accumulation are the leading causes of NAFLD. To date, no effective medicine for NAFLD has been approved by governmental agencies. Our study demonstrated that the expression of dual-specificity phosphatase 26 (Dusp26), a member of the Dusp protein family, was decreased in the liver tissue of mice with hepatic steatosis and genetically obese (ob/ob) mice. In our study, hepatic steatosis, inflammatory responses, and insulin resistance were exacerbated in liver-specific Dusp26-knockout (KO) mice but ameliorated in liver-specific Dusp26-transgenic mice induced by a high-fat diet. In addition, the degree of liver fibrosis was aggravated in high-fat high-cholesterol diet-induced Dusp26-KO mice. We further found that the binding of Dusp26 to transforming growth factor beta-activated kinase 1 (TAK1) to block the phosphorylation of TAK1 regulated the TAK1-p38/c-Jun NH2-terminal kinase signaling axis to alleviate hepatic steatosis and metabolic disturbance. Conclusion: These findings suggest that Dusp26 is a good TAK1-dependent therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Ping Ye
- Department of CardiologyThe Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jijun Liu
- Department of CardiologyThe Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Wuping Xu
- Department of NeurologyThe Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Denghai Liu
- Department of CardiologyThe Central Hospital of Wuhan, School of Medicine, Jianghan UniversityWuhanChina
| | - Xiangchao Ding
- Department of Cardiovascular SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Sheng Le
- Department of Cardiovascular SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Hao Zhang
- Department of Cardiovascular SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Shanshan Chen
- Department of Cardiovascular SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Manhua Chen
- Department of CardiologyThe Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jiahong Xia
- Department of Cardiovascular SurgeryUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
22
|
Zhou JH, Cai JJ, She ZG, Li HL. Noninvasive evaluation of nonalcoholic fatty liver disease: Current evidence and practice. World J Gastroenterol 2019; 25:1307-1326. [PMID: 30918425 PMCID: PMC6429343 DOI: 10.3748/wjg.v25.i11.1307] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 02/06/2023] Open
Abstract
With the increasing number of individuals with diabetes and obesity, nonalcoholic fatty liver disease (NAFLD) is becoming increasingly prevalent, affecting one-quarter of adults worldwide. The spectrum of NAFLD ranges from simple steatosis or nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH). NAFLD, especially NASH, may progress to fibrosis, leading to cirrhosis and hepatocellular carcinoma. NAFLD can impose a severe economic burden, and patients with NAFLD-related terminal or deteriorative liver diseases have become one of the main groups receiving liver transplantation. The increasing prevalence of NAFLD and the severe outcomes of NASH make it necessary to use effective methods to identify NAFLD. Although recognized as the gold standard, biopsy is limited by its sampling bias, poor acceptability, and severe complications, such as mortality, bleeding, and pain. Therefore, noninvasive methods are urgently needed to avoid biopsy for diagnosing NAFLD. This review discusses the current noninvasive methods for assessing NAFLD, including steatosis, NASH, and NAFLD-related fibrosis, and explores the advantages and disadvantages of measurement tools. In addition, we analyze potential noninvasive biomarkers for tracking disease processes and monitoring treatment effects, and explore effective algorithms consisting of imaging and nonimaging biomarkers for diagnosing advanced fibrosis and reducing unnecessary biopsies in clinical practice.
Collapse
Affiliation(s)
- Jiang-Hua Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Institute of Model Animal of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Jing-Jing Cai
- Department of Cardiology, The 3rd Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Institute of Model Animal of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Hong-Liang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Institute of Model Animal of Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
23
|
Yu Y, Cai J, She Z, Li H. Insights into the Epidemiology, Pathogenesis, and Therapeutics of Nonalcoholic Fatty Liver Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801585. [PMID: 30828530 PMCID: PMC6382298 DOI: 10.1002/advs.201801585] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/14/2018] [Indexed: 05/05/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease which affects ≈25% of the adult population worldwide, placing a tremendous burden on human health. The disease spectrum ranges from simple steatosis to steatohepatitis, fibrosis, and ultimately, cirrhosis and carcinoma, which are becoming leading reasons for liver transplantation. NAFLD is a complex multifactorial disease involving myriad genetic, metabolic, and environmental factors; it is closely associated with insulin resistance, metabolic syndrome, obesity, diabetes, and many other diseases. Over the past few decades, countless studies focusing on the investigation of noninvasive diagnosis, pathogenesis, and therapeutics have revealed different aspects of the mechanism and progression of NAFLD. However, effective pharmaceuticals are still in development. Here, the current epidemiology, diagnosis, animal models, pathogenesis, and treatment strategies for NAFLD are comprehensively reviewed, emphasizing the outstanding breakthroughs in the above fields and promising medications in and beyond phase II.
Collapse
Affiliation(s)
- Yao Yu
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Jingjing Cai
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Zhigang She
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| | - Hongliang Li
- Department of CardiologyRenmin Hospital of Wuhan UniversityJiefang Road 238Wuhan430060P. R. China
- Institute of Model AnimalWuhan UniversityDonghu Road 115Wuhan430071P. R. China
| |
Collapse
|
24
|
Doğan T, Yetim M, Çelik O, Kalçık M, Özcan O, Doğan İ, Erenler AK, Bekar L, Karavelioğlu Y. Investigation of mindin levels in hypertensive patients with left ventricular hypertrophy and QRS fragmentation on electrocardiography. Acta Cardiol 2018; 73:544-549. [PMID: 29265929 DOI: 10.1080/00015385.2017.1418616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Mindin was associated with diabetic nephropathy, podocyte injury, colitis, allergic asthma, liver ischaemia and reperpusion injury and ischaemic brain injury. On the other hand, it was reported as a protective factor against obesity, cardiac hypertrophy, fibrosis and remodelling. Fragmented QRS complexes (fQRS) are markers of altered ventricular depolarisation owing to a prior myocardial scar and fibrosis. In this study, we aimed to investigate mindin levels in hypertensive patients with left ventricular hypertrophy and fQRS on electrocardiography. METHODS This observational case-control study enrolled 70 (36 female) hypertensive patients with fQRS and 38 (23 female) hypertensive control patients. All patients were evaluated by transthoracic echocardiography. Mindin levels were measured by the enzyme-linked immunosorbent assay (ELISA). Clinical, echocardiographic and laboratory data were compared between patient and control groups. RESULTS There was no significant difference between patient and control groups in terms of clinical, echocardiographic and routine laboratory parameters. The mindin levels were significantly higher in the patient group than controls (11.3 (7.21-19.31) vs 4.15 (2.86-6.34); p < .001). Multiple logistic regression analyses defined increased mindin levels as an independent predictor for the presence of fQRS (Odds ratio: 1.733; p = .034). Mindin levels >6.74 predicted the presence of fQRS with a sensitifity of 84.3% and specificity of 79.9% on receiver operating characteristic (ROC) curve analysis (The area under the curve:0.889; Confidence Interval: 0.827-0.951; p < .001). CONCLUSION Mindin expressin is upregulated in hypertensive patients with fQRS complexes. In contrary to previous studies, increased mindin levels may be associated with myocardial fibrosis.
Collapse
Affiliation(s)
- Tolga Doğan
- Department of Cardiology, Hitit University Faculty of Medicine, Çorum, Turkey
| | - Mucahit Yetim
- Department of Cardiology, Hitit University Çorum Training and Research Hospital, Çorum, Turkey
| | - Oğuzhan Çelik
- Department of Cardiology, Muğla Sıtkı Koçman University Training and Research Hospital, Muğla, Turkey
| | - Macit Kalçık
- Department of Cardiology, Hitit University Faculty of Medicine, Çorum, Turkey
| | - Oğuzhan Özcan
- Department of Biochemistry, Hatay Mustafa Kemal University, Faculty of Medicine, Hatay, Turkey
| | - İbrahim Doğan
- Department of Nephrology, Hitit University Faculty of Medicine, Çorum, Turkey
| | - Ali Kemal Erenler
- Department of Emergency Medicine, Hitit University Faculty of Medicine, Çorum, Turkey
| | - Lütfü Bekar
- Department of Cardiology, Hitit University Faculty of Medicine, Çorum, Turkey
| | - Yusuf Karavelioğlu
- Department of Cardiology, Hitit University Faculty of Medicine, Çorum, Turkey
| |
Collapse
|
25
|
Dogan I, Yetim M, Dogan T, Kayadibi H, Yilmaz MB, Eser B, Kalcik M, Karavelioglu Y. Relation of serum spondin-2 levels with cardiac morphology and inflammatory parameters in hemodialysis patients. Int Urol Nephrol 2018; 50:2091-2097. [PMID: 30276603 DOI: 10.1007/s11255-018-1996-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Cardiovascular diseases are the leading causes of mortality in chronic kidney disease. Spondin-2 (SP-2), an intrinsic cardio-protective factor, prevents maladaptive remodeling. We aimed to determine the relation between serum SP-2 levels and cardiac morphology along with inflammatory parameters in hemodialysis (HD) patients. METHOD The study comprised a total of 95 patients (61 females) receiving HD treatment three times a week for at least 6 months, and a control group consisting of age and gender matched 62 subjects (34 females). SP-2 levels were determined by ELISA. Echocardiography, 24-h ambulatory blood pressure monitoring, and carotid artery intima-media thickness (CIMT) measurement were performed in all subjects. The relation of serum SP-2 levels with CIMT, echocardiographic parameters, CRP, and absolute neutrophil-to-lymphocyte count ratio (NLR) was evaluated by correlation analysis. RESULTS SP-2 levels were found to be significantly higher in the HD group than the control group (16.660 [8.719-20.938] vs. 3.988 [2.702-8.042] ng/L; P < 0.001). CIMT, CRP, and NLR were also higher in HD group (P < 0.005, P < 0.001, and P < 0.001, respectively). Significantly positive correlation was found between SP-2 and left ventricular mass, left ventricular mass index, CRP, and NLR, but no correlation was determined between SP-2 and CIMT. SP-2 was not statistically significant variable for the determination of LVH in univariate logistic regression analysis [Wald = 2.375; OR (95% CI) = 1.000 (0.999-1.000), P = 0.123]. CONCLUSION Serum SP-2 levels were higher in HD patients compared to the population with normal renal functions. The results suggest that SP-2, an uremic toxin, might be effective over a complex pathway in the inflammatory process and in the pathogenesis of cardiovascular diseases of patients under HD treatment.
Collapse
Affiliation(s)
- Ibrahim Dogan
- Department of Nephrology, School of Medicine, Hitit University, 19100, Corum, Turkey.
| | - Mucahit Yetim
- Department of Cardiology, School of Medicine, Hitit University, Corum, Turkey
| | - Tolga Dogan
- Department of Cardiology, School of Medicine, Hitit University, Corum, Turkey
| | - Huseyin Kayadibi
- Department of Biochemistry, School of Medicine, Hitit University, Corum, Turkey
| | | | - Baris Eser
- Department of Nephrology, School of Medicine, Hitit University, 19100, Corum, Turkey
| | - Macit Kalcik
- Department of Cardiology, School of Medicine, Hitit University, Corum, Turkey
| | - Yusuf Karavelioglu
- Department of Cardiology, School of Medicine, Hitit University, Corum, Turkey
| |
Collapse
|
26
|
Qin JJ, Mao W, Wang X, Sun P, Cheng D, Tian S, Zhu XY, Yang L, Huang Z, Li H. Caspase recruitment domain 6 protects against hepatic ischemia/reperfusion injury by suppressing ASK1. J Hepatol 2018; 69:1110-1122. [PMID: 29958938 DOI: 10.1016/j.jhep.2018.06.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 06/19/2018] [Accepted: 06/20/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS The hepatic injury caused by ischemia/reperfusion (I/R) insult is predominantly determined by the complex interplay of sterile inflammation and liver cell death. Caspase recruitment domain family member 6 (CARD6) was initially shown to play important roles in NF-κB activation. In our preliminary studies, CARD6 downregulation was closely related to hepatic I/R injury in liver transplantation patients and mouse models. Thus, we hypothesized that CARD6 protects against hepatic I/R injury and investigated the underlying molecular mechanisms. METHODS A partial hepatic I/R operation was performed in hepatocyte-specific Card6 knockout mice (HKO), Card6 transgenic mice with CARD6 overexpression specifically in hepatocytes (HTG), and the corresponding control mice. Hepatic histology, serum aminotransferases, inflammatory cytokines/chemokines, cell death, and inflammatory signaling were examined to assess liver damage. The molecular mechanisms of CARD6 function were explored in vivo and in vitro. RESULTS Liver injury was alleviated in Card6-HTG mice compared with control mice as shown by decreased cell death, lower serum aminotransferase levels, and reduced inflammation and infiltration, whereas Card6-HKO mice had the opposite phenotype. Mechanistically, phosphorylation of ASK1 and its downstream effectors JNK and p38 were increased in the livers of Card6-HKO mice but repressed in those of Card6-HTG mice. Furthermore, ASK1 knockdown normalized the effect of CARD6 deficiency on the activation of NF-κB, JNK and p38, while ASK1 overexpression abrogated the suppressive effect of CARD6. CARD6 was also shown to interact with ASK1. Mutant CARD6 that lacked the ability to interact with ASK1 could not inhibit ASK1 and failed to protect against hepatic I/R injury. CONCLUSIONS CARD6 is a novel protective factor against hepatic I/R injury that suppresses inflammation and liver cell death by inhibiting the ASK1 signaling pathway. LAY SUMMARY The protein CARD6 plays an important role during the process of liver blood flow restriction (ischemia) and restoration (reperfusion). By suppressing the activity of ASK1, CARD6 can protect against hepatocyte injury. Targeting CARD6 is a potential strategy for prevention and treatment of ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Juan-Juan Qin
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China; Basic Medical School, Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China
| | - Wenzhe Mao
- Basic Medical School, Wuhan University, Wuhan 430060, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China
| | - Xiaozhan Wang
- Basic Medical School, Wuhan University, Wuhan 430060, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China
| | - Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Daqing Cheng
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Song Tian
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Basic Medical School, Wuhan University, Wuhan 430060, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China
| | - Xue-Yong Zhu
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Basic Medical School, Wuhan University, Wuhan 430060, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China
| | - Ling Yang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Basic Medical School, Wuhan University, Wuhan 430060, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China
| | - Zan Huang
- College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan 430072, China.
| | - Hongliang Li
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Basic Medical School, Wuhan University, Wuhan 430060, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
27
|
Xiao XH, Wang YD, Qi XY, Wang YY, Li JY, Li H, Zhang PY, Liao HL, Li MH, Liao ZZ, Yang J, Xu CX, Wen GB, Liu JH. Zinc alpha2 glycoprotein protects against obesity-induced hepatic steatosis. Int J Obes (Lond) 2018; 42:1418-1430. [PMID: 30006580 DOI: 10.1038/s41366-018-0151-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/14/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, impaired insulin sensitivity, and chronic low-grade inflammation. Our previous studies indicated that zinc alpha2 glycoprotein (ZAG) alleviates palmitate (PA)-induced intracellular lipid accumulation in hepatocytes. This study is to further characterize the roles of ZAG on the development of hepatic steatosis, insulin resistance (IR), and inflammation. METHODS ZAG protein levels in the livers of NAFLD patients, high-fat diet (HFD)-induced or genetically (ob/ob) induced obese mice, and in PA-treated hepatocytes were determined by western blotting. C57BL/6J mice injected with an adenovirus expressing ZAG were fed HFD for indicated time to induce hepatic steatosis, IR, and inflammation, and then biomedical, histological, and metabolic analyses were conducted to identify pathologic alterations in these mice. The molecular mechanisms underlying ZAG-regulated hepatic steatosis were further explored and verified in mice and hepatocytes. RESULTS ZAG expression was decreased in NAFLD patient liver biopsy samples, obese mice livers, and PA-treated hepatocytes. Simultaneously, ZAG overexpression alleviated intracellular lipid accumulation via upregulating adiponectin and lipolytic genes (FXR, PPARα, etc.) while downregulating lipogenic genes (SREBP-1c, LXR, etc.) in obese mice as well as in cultured hepatocytes. ZAG improved insulin sensitivity and glucose tolerance via activation of IRS/AKT signaling. Moreover, ZAG significantly inhibited NF-ĸB/JNK signaling and thus resulting in suppression of obesity-associated inflammatory response in hepatocytes. CONCLUSIONS Our results revealed that ZAG could protect against NAFLD by ameliorating hepatic steatosis, IR, and inflammation.
Collapse
Affiliation(s)
- Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Ya-Di Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Xiao-Yan Qi
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Yuan-Yuan Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Jiao-Yang Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Han Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Pei-Ying Zhang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Hai-Lin Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Mei-Hua Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Zhe-Zhen Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Jing Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Can-Xin Xu
- Department of Pathology & Immunology, Washington University in, St. Louis, MO, 63110, USA
| | - Ge-Bo Wen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Jiang-Hua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China.
| |
Collapse
|
28
|
Zhang C, Qin JJ, Gong FH, Tong JJ, Cheng WL, Wang H, Zhang Y, Zhu X, She ZG, Xia H, Zhu LH. Mindin deficiency in macrophages protects against foam cell formation and atherosclerosis by targeting LXR-β. Clin Sci (Lond) 2018; 132:1199-1213. [PMID: 29695588 DOI: 10.1042/cs20180033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Mindin, which is a highly conserved extracellular matrix protein, has been documented to play pivotal roles in regulating angiogenesis, inflammatory processes, and immune responses. The aim of the present study was to assess whether mindin contributes to the development of atherosclerosis. A significant up-regulation of Mindin expression was observed in the serum, arteries and atheromatous plaques of ApoE-/- mice after high-fat diet treatment. Mindin-/-ApoE-/- mice and macrophage-specific mindin overexpression in ApoE-/- mice (Lyz2-mindin-TG) were generated to evaluate the effect of mindin on the development of atherosclerosis. The Mindin-/-ApoE-/- mice exhibited significantly ameliorated atherosclerotic burdens in the entire aorta and aortic root and increased atherosclerotic plaque stability. Moreover, bone marrow transplantation further demonstrated that mindin deficiency in macrophages was largely responsible for the alleviated atherogenesis. The Lyz2-mindin-TG mice exhibited the opposite phenotype. Mindin deficiency enhanced foam cell formation by increasing the expression of cholesterol effectors, including ABCA1 and ABCG1. The mechanistic study indicated that mindin ablation promoted LXR-β expression via a direct interaction. Importantly, LXR-β inhibition largely reversed the ameliorating effect of mindin deficiency on foam cell formation and ABCA1 and ABCG1 expression. The present study demonstrated that mindin deficiency serves as a novel mediator that protects against foam cell formation and atherosclerosis by directly interacting with LXR-β.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Fu-Han Gong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Jing-Jing Tong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Wen-Lin Cheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Haiping Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Xueyong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, P.R. China
| |
Collapse
|
29
|
p38α MAPK antagonizing JNK to control the hepatic fat accumulation in pediatric patients onset intestinal failure. Cell Death Dis 2017; 8:e3110. [PMID: 29022907 PMCID: PMC5682685 DOI: 10.1038/cddis.2017.523] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 08/23/2017] [Accepted: 09/07/2017] [Indexed: 12/30/2022]
Abstract
The p38α mitogen-activated protein kinase (MAPK) has been related to gluconeogenesis and lipid metabolism. However, the roles and related mechanisms of p38α MAPK in intestinal failure (IF)-associated liver steatosis remained poor understood. Here, our experimental evidence suggested that p38α MAPK significantly suppressed the fat accumulation in livers of IF patients mainly through two mechanisms. On the one hand, p38α MAPK increased hepatic bile acid (BA) synthesis by upregulating the expression of the rate-limiting enzyme cholesterol 7-α-hydroxylase (CYP7A1) and peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), which in turn activated the transcription of the CYP7A1. On the other hand, p38α MAPK promoted fatty acid (FA) β-oxidation via upregulating peroxisome proliferator-activated receptor alpha (PPARα) and its transcriptional target genes carnitine palmitoyltransferase 1A (CPT1A) and peroxisomal acyl-coenzyme aoxidase 1 (ACOX1). Dual luciferase assays indicated that p38α MAPK increased the transcription of PPARα, PGC-1α and CYP7A1 by upregulating their promoters’ activities. In addition, in vitro and in vivo assays indicated p38α MAPK negatively regulates the hepatic steatosis by controlling JNK activation. In conculsion, our findings demonstrate that hepatic p38α MAPK functions as a negative regulator of liver steatosis in maintaining BA synthesis and FAO by antagonizing the c-Jun N-terminal kinase (JNK).
Collapse
|
30
|
Wang LF, Liu YS, Yang B, Li P, Cheng XS, Xiao CX, Liu JJ, Li S, Ren JL, Guleng B. The extracellular matrix protein mindin attenuates colon cancer progression by blocking angiogenesis via Egr-1-mediated regulation. Oncogene 2017; 37:601-615. [PMID: 28991232 DOI: 10.1038/onc.2017.359] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 08/15/2017] [Accepted: 08/19/2017] [Indexed: 12/13/2022]
Abstract
Mindin, a secreted, highly conserved extracellular matrix (ECM) protein, exerts a broad spectrum of effects on the innate immune system. However, its function in colorectal cancer (CRC) progression is not well established, and its upstream regulation mechanisms remain unclear. Contrary to previous reports, this study used two different enzyme-linked immunosorbent assay (ELISA) kits to show that the serum level of mindin was significantly decreased in CRC patients and that this decreased level is more significantly associated with the early stages of the disease. To explore the regulation of mindin, we used a bioinformatics approach to predict potential transcription factors and determined that early growth response factor (Egr)-1 directly regulates mindin expression at the transcriptional level using dual luciferase, chromatin immunoprecipitation (ChIP) DNA and electrophoretic mobility shift assay (EMSA) methods. Egr-1 regulates mindin mRNA and protein expression in CRC cells, and the protein expression of both Egr-1 and mindin was significantly decreased in tumor lesions of patients compared with adjacent control tissues. Mindin is essential for Egr-1-mediated inhibition of endothelial cell tube formation, and mindin inhibits endotheliocyte proliferation, migration and angiogenic sprouts in vitro. Overexpression of mindin suppressed xenograft tumor growth by blocking angiogenesis instead of directly suppressing CRC cell proliferation. Mechanically, mindin inhibits the hypoxia-induced HIF-1a and VEGFA protein expression in CRC cells and the phosphorylation of VEGFR-2 in endothelial cells. The results suggest that the serum level of mindin can be used as a novel biomarker for early detection of CRC and that the Egr-1/mindin axis is a potential therapeutic target for the inhibition of angiogenesis in CRC development.
Collapse
Affiliation(s)
- L-F Wang
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - Y-S Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - B Yang
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - P Li
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China.,Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - X-S Cheng
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - C-X Xiao
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - J-J Liu
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - S Li
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China
| | - J-L Ren
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China
| | - B Guleng
- Department of Gastroenterology, Zhongshan Hospital affiliated to Xiamen University, Xiamen, China.,Faculty of Clinical Medicine, Medical College of Xiamen University, Xiamen, China.,State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, China
| |
Collapse
|
31
|
Zhang QY, Zhao LP, Tian XX, Yan CH, Li Y, Liu YX, Wang PX, Zhang XJ, Han YL. The novel intracellular protein CREG inhibits hepatic steatosis, obesity, and insulin resistance. Hepatology 2017; 66:834-854. [PMID: 28508477 DOI: 10.1002/hep.29257] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 04/02/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022]
Abstract
UNLABELLED Cellular repressor of E1A-stimulated genes (CREG), a novel cellular glycoprotein, has been identified as a suppressor of various cardiovascular diseases because of its capacity to reduce hyperplasia, maintain vascular homeostasis, and promote endothelial restoration. However, the effects and mechanism of CREG in metabolic disorder and hepatic steatosis remain unknown. Here, we report that hepatocyte-specific CREG deletion dramatically exacerbates high-fat diet and leptin deficiency-induced (ob/ob) adverse effects such as obesity, hepatic steatosis, and metabolic disorders, whereas a beneficial effect is conferred by CREG overexpression. Additional experiments demonstrated that c-Jun N-terminal kinase 1 (JNK1) but not JNK2 is largely responsible for the protective effect of CREG on the aforementioned pathologies. Notably, JNK1 inhibition strongly prevents the adverse effects of CREG deletion on steatosis and related metabolic disorders. Mechanistically, CREG interacts directly with apoptosis signal-regulating kinase 1 (ASK1) and inhibits its phosphorylation, thereby blocking the downstream MKK4/7-JNK1 signaling pathway and leading to significantly alleviated obesity, insulin resistance, and hepatic steatosis. Importantly, dramatically reduced CREG expression and hyperactivated JNK1 signaling was observed in the livers of nonalcoholic fatty liver disease (NAFLD) patients, suggesting that CREG might be a promising therapeutic target for NAFLD and related metabolic diseases. CONCLUSION The results of our study provides evidence that CREG is a robust suppressor of hepatic steatosis and metabolic disorders through its direct interaction with ASK1 and the resultant inactivation of ASK1-JNK1 signaling. This study offers insights into NAFLD pathogenesis and its complicated pathologies, such as obesity and insulin resistance, and paves the way for disease treatment through targeting CREG. (Hepatology 2017;66:834-854).
Collapse
Affiliation(s)
- Quan-Yu Zhang
- Graduate School of Third Military Medical University, Chongqing, China.,Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Ling-Ping Zhao
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Xiao-Xiang Tian
- Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Cheng-Hui Yan
- Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yang Li
- Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Yan-Xia Liu
- Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| | - Pi-Xiao Wang
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Ya-Ling Han
- Department of Cardiology, General Hospital of Shenyang Military Region, Shenyang, China
| |
Collapse
|
32
|
Yan FJ, Zhang XJ, Wang WX, Ji YX, Wang PX, Yang Y, Gong J, Shen LJ, Zhu XY, Huang Z, Li H. The E3 ligase tripartite motif 8 targets TAK1 to promote insulin resistance and steatohepatitis. Hepatology 2017; 65:1492-1511. [PMID: 27981609 DOI: 10.1002/hep.28971] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/22/2016] [Indexed: 12/20/2022]
Abstract
UNLABELLED Tripartite motif 8 (TRIM8), an E3 ligase ubiquitously expressed in various cells, is closely involved in innate immunity. However, its role in nonalcoholic steatohepatitis is largely unknown. Here, we report evidence that TRIM8 is a robust enhancer of steatohepatitis and its complications induced by a high-fat diet or a genetic deficiency (ob/ob). Using gain-of-function and loss-of-function approaches, we observed dramatic exacerbation of insulin resistance, hepatic steatosis, inflammation, and fibrosis by hepatocyte-specific TRIM8 overexpression, whereas deletion or down-regulation of TRIM8 in hepatocytes led to a completely opposite phenotype. Furthermore, investigations of the underlying mechanisms revealed that TRIM8 directly binds to and ubiquitinates transforming growth factor-beta-activated kinase 1, thus promoting its phosphorylation and the activation of downstream c-Jun N-terminal kinase/p38 and nuclear factor κB signaling. Importantly, the participation of TRIM8 in human nonalcoholic fatty liver disease and nonalcoholic steatohepatitis was verified on the basis of its dramatically increased expression in the livers of these patients, suggesting a promising development of TRIM8 disturbance for the treatment of nonalcoholic steatohepatitis-related metabolic disorders. CONCLUSION The E3 ligase TRIM8 is a potent regulator that exacerbates steatohepatitis and metabolic disorders dependent on its binding and ubiquitinating capacity on transforming growth factor-beta-activated kinase 1. (Hepatology 2017;65:1492-1511).
Collapse
Affiliation(s)
- Feng-Juan Yan
- College of Life Sciences, Wuhan University, Wuhan, China.,School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Wen-Xin Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Pi-Xiao Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yang Yang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Gong
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Li-Jun Shen
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zan Huang
- College of Life Sciences, Wuhan University, Wuhan, China.,School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hongliang Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Institute of Model Animals, Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Kong Q, Zhang H, Zhao T, Zhang W, Yan M, Dong X, Li P. Tangshen formula attenuates hepatic steatosis by inhibiting hepatic lipogenesis and augmenting fatty acid oxidation in db/db mice. Int J Mol Med 2016; 38:1715-1726. [PMID: 27840945 PMCID: PMC5117754 DOI: 10.3892/ijmm.2016.2799] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 11/03/2016] [Indexed: 12/24/2022] Open
Abstract
Tangshen formula (TSF), a well-prescribed traditional Chinese formula, has been used in the treatment of diabetic nephropathy. However, whether TSF ameliorates dyslipidemia and liver injury associated with diabetes remains unclear. In this study, we examined the effects of TSF on lipid profiles and hepatic steatosis in db/db mice. For this purpose, 8‑week-old db/db mice were treated with TSF or saline for 12 weeks via gavage and db/m mice were used as controls. Body weight and blood glucose levels were monitored weekly and bi-weekly, respectively. Blood samples were obtained for the analysis of lipids and enzymes related to hepatic function, and liver tissues were analyzed by histology, immunohistochemistry and molecular examination. The results revealed that TSF markedly reduced body weight, liver index [liver/body weight (LW/BW)] and improved lipid profiles, hepatic function and steatosis in db/db mice. TSF induced the phosphoralation of AMP-activated protein kinase and inhibited the activity of sterol regulatory element-binding protein 1 together with the inhibition of the expression of genes involved in de novo lipogenesis (DNL) and gluconeogenesis, such as fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), stearoyl CoA desaturase 1 (SCD1), glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1). Additionally, the silent mating type information regulation 2 homolog 1 (Sirt1)/peroxisome proliferator-activated receptor α (PPARα)/malonyl-CoA decarboxylase (MLYCD) cascade was potently activated by TSF in the liver and skeletal muscle of db/db mice, which led to enhanced fatty acid oxidation. These findings demonstrated that TSF attenuated hepatic fat accumulation and steatosis in db/db mice by inhibiting lipogenesis and augmenting fatty acid oxidation.
Collapse
Affiliation(s)
- Qin Kong
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| | - Haojun Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Tingting Zhao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Weiku Zhang
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Meihua Yan
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Xi Dong
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, P.R. China
| | - Ping Li
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, P.R. China
| |
Collapse
|
34
|
Xie L, Wang PX, Zhang P, Zhang XJ, Zhao GN, Wang A, Guo J, Zhu X, Zhang Q, Li H. DKK3 expression in hepatocytes defines susceptibility to liver steatosis and obesity. J Hepatol 2016; 65:113-124. [PMID: 27016281 DOI: 10.1016/j.jhep.2016.03.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/09/2016] [Accepted: 03/13/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Dickkopf-3 (DKK3), a protein belonging to the DKK family, has been extensively investigated in the context of cancer, including liver cancer. However, the role of DKK3 in hepatic steatosis and related metabolic disorders remains largely unexplored. METHODS We detected the expression of DKK3 in the fatty livers of NAFLD patients and of obese mice and investigated the function of DKK3 in hepatic steatosis and related metabolic disorders by using hepatocyte-specific DKK3 deficiency or overexpression obese mice induced by high fat diet (HFD) or genetic defect (ob/ob). The molecular mechanisms underlying DKK3-regulated hepatic steatosis were further explored and verified in mice. RESULTS DKK3 expression was significantly decreased in the livers of NAFLD patients and of obese mice as well as in cultured hepatocytes stimulated with palmitate. Further investigation indicated that specific overexpression of DKK3 in hepatocytes enhanced insulin sensitivity and glucose tolerance, reduced the inflammatory response, and ameliorated the imbalance of lipid metabolism in response to HFD or genetic defects. In contrast, DKK3 deficiency in hepatocytes led to an almost complete reversal of these pathologies. Mechanistically, DKK3 combined with Apoptosis signal-regulating kinase 1 (ASK1) under palmitate stimulation, and thus inhibited the activation of the downstream P38/JNK pathway. Importantly, dominant-negative ASK1 blocked the accelerated effects of DKK3 deficiency, while the constitutively active form of ASK1 overcame the inhibitory effects of DKK3 overexpression on HFD-induced metabolic disorders in vivo. CONCLUSION DKK3 functions as a negative regulator of insulin resistance, hepatic steatosis, and associated inflammatory responses, which depends on its inhibitory regulation of ASK1 activity. LAY SUMMARY DKK3 expression is decreased in the non-alcoholic fatty liver of humans and mice. Adding DKK3 expression alleviates fatty liver in mice by inhibiting ASK1 activity.
Collapse
Affiliation(s)
- Lanfeng Xie
- Department of Hepatology and Infection, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China
| | - Pi-Xiao Wang
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Peng Zhang
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xiao-Jing Zhang
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China
| | - Guang-Nian Zhao
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Aibing Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Junhong Guo
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xueyong Zhu
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Qin Zhang
- Department of Hepatology and Infection, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| | - Hongliang Li
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430071, China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
35
|
Xiang M, Wang PX, Wang AB, Zhang XJ, Zhang Y, Zhang P, Mei FH, Chen MH, Li H. Targeting hepatic TRAF1-ASK1 signaling to improve inflammation, insulin resistance, and hepatic steatosis. J Hepatol 2016; 64:1365-77. [PMID: 26860405 DOI: 10.1016/j.jhep.2016.02.002] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/14/2016] [Accepted: 02/01/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Tumor necrosis factor receptor-associated factor 1 (TRAF1) is an important adapter protein that is largely implicated in molecular events regulating immunity/inflammation and cell death. Although inflammation is closely related to and forms a vicious circle with insulin dysfunction and hepatic lipid accumulation, the role of TRAF1 in hepatic steatosis and the related metabolic disorders remains unclear. METHODS The participation of TRAF1 in the initiation and progression of hepatic steatosis was evaluated in high fat diet (HFD)-induced and genetic obesity. Mice with global TRAF1 knockout or liver-specific TRAF1 overexpression were employed to investigate the role of TRAF1 in insulin resistance, inflammation, and hepatic steatosis based on various phenotypic examinations. Molecular mechanisms underlying TRAF1-regulated hepatic steatosis were further explored in vivo and in vitro. RESULTS TRAF1 expression was significantly upregulated in the livers of NAFLD patients and obese mice and in palmitate-treated hepatocytes. In response to HFD administration or in ob/ob mice, TRAF1 deficiency was hepatoprotective, whereas the overexpression of TRAF1 in hepatocytes contributed to the pathological development of insulin resistance, inflammatory response and hepatic steatosis. Mechanistically, hepatocyte TRAF1 promotes hepatic steatosis through enhancing the activation of ASK1-mediated P38/JNK cascades, as evidenced by the fact that ASK1 inhibition abolished the exacerbated effect of TRAF1 on insulin dysfunction, inflammation, and hepatic lipid accumulation. CONCLUSIONS TRAF1 functions as a positive regulator of insulin resistance, inflammation, and hepatic steatosis dependent on the activation of ASK1-P38/JNK axis.
Collapse
Affiliation(s)
- Mei Xiang
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Ai-Bing Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Xiao-Jing Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Yaxing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Fang-Hua Mei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Man-Hua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Wuhan, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
36
|
Luo P, Wang PX, Li ZZ, Zhang XJ, Jiang X, Gong J, Qin JJ, Guo J, Zhu X, Yang S, Li H. Hepatic Oncostatin M Receptor β Regulates Obesity-Induced Steatosis and Insulin Resistance. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1278-92. [PMID: 26976243 DOI: 10.1016/j.ajpath.2015.12.028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/03/2015] [Accepted: 12/31/2015] [Indexed: 01/07/2023]
Abstract
The liver is an essential insulin-responsive organ that is critical for maintaining glucose homeostasis and lipid metabolism. Oncostatin M receptor β chain (OSMRβ) is implicated in adipose tissue- and immune cell-mediated metabolic regulation. However, the role of hepatocyte-derived OSMRβ in metabolic disorders remains unclear. Here, we report on the central role of OSMRβ in the protection against obesity and deregulation of glucose and lipids. We observed significantly varied expression levels of OSMRβ in hepatic tissues in both human samples and mouse models of nonalcoholic fatty liver disease. Mice lacking either whole-body or hepatic OSMRβ displayed exacerbated diet-induced insulin resistance, hepatic steatosis, and inflammation, both in diet-induced and genetically (ob/ob) obese mice. These adverse effects were markedly attenuated by hepatocyte-specific overexpression of OSMRβ. Mechanistically, we showed that OSMRβ phosphorylates and activates the Janus kinase 2 (JAK2)/STAT3 signaling pathway in the liver. More importantly, the liver-restricted overexpression of STAT3 rescued glucose tolerance and ameliorated hepatic steatosis and inflammation in OSMRβ knockout mice, whereas OSMRβ overexpression failed to protect against hepatic steatosis, insulin resistance, and hepatic inflammation in STAT3-deficient mice. Thus, activation of STAT3 is both sufficient and required to produce OSMRβ-mediated beneficial effects. In conclusion, hepatic OSMRβ expression alleviates obesity-induced hepatic insulin resistance and steatosis through the activation of JAK2/STAT3 signaling cascades.
Collapse
Affiliation(s)
- Pengcheng Luo
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Huangshi Central Hospital, Hubei Polytechnic University, Huangshi, China
| | - Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Zuo-Zhi Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Jing Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xi Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Jun Gong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China; College of Life Sciences, Wuhan University, Wuhan, China
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Junhong Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Xueyong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Sijun Yang
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China.
| |
Collapse
|
37
|
Wang PX, Zhang XJ, Luo P, Jiang X, Zhang P, Guo J, Zhao GN, Zhu X, Zhang Y, Yang S, Li H. Hepatocyte TRAF3 promotes liver steatosis and systemic insulin resistance through targeting TAK1-dependent signalling. Nat Commun 2016; 7:10592. [PMID: 26882989 PMCID: PMC4757796 DOI: 10.1038/ncomms10592] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, insulin resistance and a systemic pro-inflammatory response. Here we show that tumour necrosis factor receptor-associated factor 3 (TRAF3) is upregulated in mouse and human livers with hepatic steatosis. After 24 weeks on a high-fat diet (HFD), obesity, insulin resistance, hepatic steatosis and inflammatory responses are significantly ameliorated in liver-specific TRAF3-knockout mice, but exacerbated in transgenic mice overexpressing TRAF3 in hepatocytes. The detrimental effects of TRAF3 on hepatic steatosis and related pathologies are confirmed in ob/ob mice. We further show that in response to HFD, hepatocyte TRAF3 binds to TGF-β-activated kinase 1 (TAK1) to induce TAK1 ubiquitination and subsequent autophosphorylation, thereby enhancing the activation of downstream IKKβ-NF-κB and MKK-JNK-IRS1(307) signalling cascades, while disrupting AKT-GSK3β/FOXO1 signalling. The TRAF3-TAK1 interaction and TAK1 ubiquitination are indispensable for TRAF3-regulated hepatic steatosis. In conclusion, hepatocyte TRAF3 promotes HFD-induced or genetic hepatic steatosis in a TAK1-dependent manner.
Collapse
Affiliation(s)
- Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Xiao-Jing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China.,State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Pengcheng Luo
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Huangshi Central Hospital, Hubei Polytechnic University, Huangshi 435000, China
| | - Xi Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Junhong Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Guang-Nian Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Xueyong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Yan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Sijun Yang
- Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan 430060, China
| |
Collapse
|
38
|
Brahma Naidu P, Uddandrao VVS, Ravindar Naik R, Suresh P, Meriga B, Begum MS, Pandiyan R, Saravanan G. Ameliorative potential of gingerol: Promising modulation of inflammatory factors and lipid marker enzymes expressions in HFD induced obesity in rats. Mol Cell Endocrinol 2016; 419:139-147. [PMID: 26493465 DOI: 10.1016/j.mce.2015.10.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/10/2015] [Accepted: 10/11/2015] [Indexed: 01/12/2023]
Abstract
Obesity, generally linked to hyperlipidemia, has been occurring of late with distressing alarm and has now become a global phenomenon casting a huge economic burden on the health care system of countries around the world. The present study investigated the effects of gingerol over 30 days on the changes in HFD-induced obese rats in marker enzymes of lipid metabolism such as fatty-acid synthase (FAS), Acetyl CoA Carboxylase (ACC), Carnitine Palmitoyl Transferase-1(CPT-1), HMG co-A Reductase (HMGR), Lecithin Choline Acyl Transferase (LCAT) and Lipoprotein Lipase (LPL) and inflammatory markers (TNF-α and IL-6). The rats were treated orally with gingerol (75 mg kg(-1)) once daily for 30 days with a lorcaserin-treated group (10 mg kg(-1)) included for comparison. Changes in body weight, glucose, insulin resistance and expressions of lipid marker enzymes and inflammatory markers in tissues were observed in experimental rats. The administration of gingerol resulted in a significant reduction in body weight gain, glucose and insulin levels, and insulin resistance, which altered the activity, expressions of lipid marker enzymes and inflammatory markers. It showed that gingerol had significantly altered these parameters when compared with HFD control rats. This study confirms that gingerol prevents HFD-induced hyperlipidemia by modulating the expression of enzymes important to cholesterol metabolism.
Collapse
Affiliation(s)
- Parim Brahma Naidu
- Department of Biochemistry, Animal Physiology & Biochemistry Lab, Sri Venkateswara University, Tirupati, 517502, India
| | - V V Sathibabu Uddandrao
- Department of Biochemistry, Centre for Biological Sciences, K.S. Rangasamy College of Arts and Science, Thokkavadi, Tiruchengode, 637215, Tamil Nadu, India
| | - Ramavat Ravindar Naik
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition (ICMR-New Delhi), Hydrabad, Andhrapradesh, India
| | - Pothani Suresh
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition (ICMR-New Delhi), Hydrabad, Andhrapradesh, India
| | - Balaji Meriga
- Department of Biochemistry, Animal Physiology & Biochemistry Lab, Sri Venkateswara University, Tirupati, 517502, India
| | - Mustapha Shabana Begum
- Department of Biochemistry, Muthayammal College of Arts and Science, Rasipuram, Tamil Nadu, 637408, India
| | - Rajesh Pandiyan
- Department of Biochemistry, Centre for Biological Sciences, K.S. Rangasamy College of Arts and Science, Thokkavadi, Tiruchengode, 637215, Tamil Nadu, India
| | - Ganapathy Saravanan
- Department of Biochemistry, Centre for Biological Sciences, K.S. Rangasamy College of Arts and Science, Thokkavadi, Tiruchengode, 637215, Tamil Nadu, India.
| |
Collapse
|
39
|
Hu J, Zhu XH, Zhang XJ, Wang PX, Zhang R, Zhang P, Zhao GN, Gao L, Zhang XF, Tian S, Li H. Targeting TRAF3 signaling protects against hepatic ischemia/reperfusions injury. J Hepatol 2016; 64:146-59. [PMID: 26334576 DOI: 10.1016/j.jhep.2015.08.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 08/01/2015] [Accepted: 08/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The hallmarks of hepatic ischemia/reperfusion (I/R) injury, a common clinical problem that occurs during liver surgical procedures, include severe cell death and inflammatory responses that contribute to early graft failure and a higher incidence of organ rejection. Unfortunately, effective therapeutic strategies are limited. Tumor necrosis factor receptor (TNFR)-associated factor (TRAF) 3 transduces apoptosis and/or inflammation-related signaling pathways to regulate cell survival and cytokine production. However, the role of TRAF3 in hepatic I/R-induced liver damage remains unknown. METHODS Hepatocyte- or myeloid cell-specific TRAF3 knockdown or transgenic mice were subjected to an I/R model in vivo, and in vitro experiments were performed by treating primary hepatocytes from these mice with hypoxia/reoxygenation stimulation. The function of TRAF3 in I/R-induced liver damage and the potential underlying mechanisms were investigated through various phenotypic analyses and biological approaches. RESULTS Hepatocyte-specific, but not myeloid cell-specific, TRAF3 deficiency reduced cell death, inflammatory cell infiltration, and cytokine production in both in vivo and in vitro hepatic I/R models, whereas hepatic TRAF3 overexpression resulted in the opposite effects. Mechanistically, TRAF3 directly binds to TAK1, which enhances the activation of the downstream NF-κB and JNK pathways. Importantly, inhibition of TAK1 almost completely reversed the TRAF3 overexpression-mediated exacerbation of I/R injury. CONCLUSIONS TRAF3 is a novel hepatic I/R mediator that promotes liver damage and inflammation via TAK1-dependent activation of the JNK and NF-κB pathways. Inhibition of hepatic TRAF3 may represent a promising approach to protect the liver against I/R injury-related diseases.
Collapse
Affiliation(s)
- Junfei Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Xue-Hai Zhu
- Department of Thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Jing Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Ran Zhang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Guang-Nian Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Lu Gao
- Department of Cardiology, Institute of Cardiovascular Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Fei Zhang
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Animal Experiment Center/Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China.
| |
Collapse
|
40
|
Sun P, Zhang P, Wang PX, Zhu LH, Du Y, Tian S, Zhu X, Li H. Mindin deficiency protects the liver against ischemia/reperfusion injury. J Hepatol 2015; 63:1198-211. [PMID: 26165142 DOI: 10.1016/j.jhep.2015.06.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Hepatic ischemia/reperfusion (I/R) injury often occurs during liver surgery and may cause liver failure. Our previous studies revealed that Mindin is involved in the pathogenesis of ischemic stroke. However, the function of Mindin in hepatic I/R injury remains unknown. METHODS Partial hepatic warm ischemia was induced in parallel in global Mindin knockout mice (Mindin KO), hepatocyte-specific Mindin knockdown mice, hepatocyte-specific Mindin transgenic mice (Mindin TG), myeloid cell-specific Mindin TG mice (LysM-Mindin TG), and their corresponding controls, followed by reperfusion. Hepatic histology, serum aminotransferase, inflammatory cytokines, and hepatocyte apoptosis and proliferation were examined to assess liver injury. The molecular mechanisms of Mindin function were explored in vivo and in vitro. RESULTS Mindin KO and hepatocyte-specific Mindin knockdown mice exhibited less liver damage than controls, with smaller necrotic areas and lower serum transaminase levels. Mindin deficiency significantly suppressed inflammatory cell infiltration, cytokine and chemokine production, and hepatocyte apoptosis, but increased hepatocyte proliferation following hepatic I/R injury. In contrast, the opposite pathological and biochemical changes were observed in hepatocyte-specific Mindin TG mice, whereas no significant changes in liver damage were found in LysM-Mindin TG mice compared to non-transgenic controls. Mechanistically, Akt signaling was activated in livers of Mindin KO mice but was suppressed in Mindin TG mice. Most importantly, Akt inhibitor treatment blocked the protective effect of Mindin deficiency on hepatic I/R injury. CONCLUSIONS Mindin is a novel modulator of hepatic I/R injury through regulating inflammatory responses, as well as hepatocyte apoptosis and proliferation via inactivation of the Akt signaling pathway.
Collapse
Affiliation(s)
- Peng Sun
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Peng Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Pi-Xiao Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Li-Hua Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yibao Du
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Song Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Xueyong Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China.
| |
Collapse
|
41
|
Zhang XJ, Zhang P, Li H. Interferon regulatory factor signalings in cardiometabolic diseases. Hypertension 2015; 66:222-47. [PMID: 26077571 DOI: 10.1161/hypertensionaha.115.04898] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/14/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Xiao-Jing Zhang
- From the Department of Cardiology, Renmin Hospital (X.-J.Z., P.Z., H.L.) and Cardiovascular Research Institute (X.-J.Z., P.Z., H.L.), Wuhan University, Wuhan, China; and State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, PR China (X.-J.Z.)
| | - Peng Zhang
- From the Department of Cardiology, Renmin Hospital (X.-J.Z., P.Z., H.L.) and Cardiovascular Research Institute (X.-J.Z., P.Z., H.L.), Wuhan University, Wuhan, China; and State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, PR China (X.-J.Z.)
| | - Hongliang Li
- From the Department of Cardiology, Renmin Hospital (X.-J.Z., P.Z., H.L.) and Cardiovascular Research Institute (X.-J.Z., P.Z., H.L.), Wuhan University, Wuhan, China; and State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, PR China (X.-J.Z.).
| |
Collapse
|
42
|
Mindin regulates vascular smooth muscle cell phenotype and prevents neointima formation. Clin Sci (Lond) 2015; 129:129-45. [PMID: 25751394 DOI: 10.1042/cs20140679] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In the present study, using diverse in vitro and in vivo models, we revealed that mindin is a novel modulator of VSMC phenotype and neointima formation in an AKT-dependent manner in response to vascular injury.
Collapse
|
43
|
Seo KI, Lee J, Choi RY, Lee HI, Lee JH, Jeong YK, Kim MJ, Lee MK. Anti-obesity and anti-insulin resistance effects of tomato vinegar beverage in diet-induced obese mice. Food Funct 2014; 5:1579-1586. [PMID: 24867606 DOI: 10.1039/c4fo00135d] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2025]
Abstract
This study investigated the mechanism of processed tomato vinegar beverage (TVB)-mediated anti-obesity and anti-insulin resistance effects in high-fat diet (HF)-induced obese mice. Oral administration of TVB (14 mL kg(-1) body weight) to HF-fed mice for 6 weeks effectively reduced the body and visceral fat weight and significantly lowered plasma free fatty acid, triglyceride and hepatic triglyceride levels. TVB significantly increased fecal triglyceride excretion, both phosphorylated AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) and peroxisome proliferator-activated receptor (PPAR)α protein levels in the liver, which were associated with increased fatty acid β-oxidation and carnitine palmitoyltransferase activities in HF-fed mice. TVB improved glucose tolerance, hyperinsulinemia and HOMA-IR levels in the HF + TVB group compared to the HF group. Additionally, TVB significantly increased glucokinase activity and decreased glucose-6-phosphatase activity in the liver, which enhanced glucose metabolism in obese mice. These results suggest that TVB prevents visceral obesity and insulin resistance via AMPK/PPARα-mediated fatty acid and glucose oxidation.
Collapse
Affiliation(s)
- Kwon-Il Seo
- Department of Food and Nutrition, Sunchon National University, Suncheon 540-950, Korea.
| | | | | | | | | | | | | | | |
Collapse
|