1
|
Zhao J, Duan L, Li J, Yao C, Wang G, Mi J, Yu Y, Ding L, Zhao Y, Yan G, Li J, Zhao Z, Wang X, Li M. New insights into the interplay between autophagy, gut microbiota and insulin resistance in metabolic syndrome. Biomed Pharmacother 2024; 176:116807. [PMID: 38795644 DOI: 10.1016/j.biopha.2024.116807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024] Open
Abstract
Metabolic syndrome (MetS) is a widespread and multifactorial disorder, and the study of its pathogenesis and treatment remains challenging. Autophagy, an intracellular degradation system that maintains cellular renewal and homeostasis, is essential for maintaining antimicrobial defense, preserving epithelial barrier integrity, promoting mucosal immune response, maintaining intestinal homeostasis, and regulating gut microbiota and microbial metabolites. Dysfunctional autophagy is implicated in the pathological mechanisms of MetS, involving insulin resistance (IR), chronic inflammation, oxidative stress, and endoplasmic reticulum (ER) stress, with IR being a predominant feature. The study of autophagy represents a valuable field of research with significant clinical implications for identifying autophagy-related signals, pathways, mechanisms, and treatment options for MetS. Given the multifactorial etiology and various potential risk factors, it is imperative to explore the interplay between autophagy and gut microbiota in MetS more thoroughly. This will facilitate the elucidation of new mechanisms underlying the crosstalk among autophagy, gut microbiota, and MetS, thereby providing new insights into the diagnosis and treatment of MetS.
Collapse
Affiliation(s)
- Jinyue Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Liyun Duan
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jiarui Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Chensi Yao
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoqiang Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jia Mi
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yongjiang Yu
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Lu Ding
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yunyun Zhao
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Guanchi Yan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jing Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Zhixuan Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xiuge Wang
- The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130021, China.
| | - Min Li
- Molecular Biology Laboratory, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
2
|
Zhao C, Song Y, Zhang G, Zhang K, Yin S, Ji J. Multi-omics analysis identifies sex-specific hepatic protein-metabolite networks in yellow catfish (Pelteobagrus fulvidraco) exposed to chronic hypoxia. Int J Biol Macromol 2024; 268:131892. [PMID: 38677698 DOI: 10.1016/j.ijbiomac.2024.131892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
Hypoxia disrupts the endocrine system of teleosts. The liver plays important roles in the endocrine system, energy storage, and metabolic processes. The aim of this study was to investigate the sex-specific hepatic response of yellow catfish under chronic hypoxia at the multi-omics level. Common hepatic responses in both sexes included the HIF-1 signaling pathway, glycolysis/gluconeogenesis, and steroid biosynthesis. Hypoxia dysregulated primary bile acid biosynthesis, lipid metabolism, and vitellogenin levels in female fish. Endoplasmic reticulum function in females also tended to be disrupted by hypoxia, as evidenced by significantly enriched pathways, including ribosome, protein processing in the endoplasmic reticulum, and RNA degradation. Other pathways, including the TCA cycle, oxidative phosphorylation, and Parkinson's and Huntington's disease, were highly enriched by hypoxia in male fish, suggesting that mitochondrial function was dysregulated. In both sexes of yellow catfish, the cell cycle was arrested and apoptosis was inhibited under chronic hypoxia. Multi-omics suggested that SLC2A5, CD209, LGMN, and NEDD8 served as sex-specific markers in these fish under chronic hypoxia. Our results provide insights into hepatic adaptation to chronic hypoxia and facilitate our understanding of sex-specific responses in fish.
Collapse
Affiliation(s)
- Cheng Zhao
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, China
| | - Yufeng Song
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, China
| | - Guosong Zhang
- School of Agriculture and Bioengineering, Heze University, Heze, Shandong, China
| | - Kai Zhang
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, China
| | - Shaowu Yin
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, China.
| | - Jie Ji
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, China; Co-Innovation Center for Marine Bio-Industry Technology, Lian Yungang, Jiangsu, China.
| |
Collapse
|
3
|
Solanki S, Shah YM. Hypoxia-Induced Signaling in Gut and Liver Pathobiology. ANNUAL REVIEW OF PATHOLOGY 2024; 19:291-317. [PMID: 37832943 DOI: 10.1146/annurev-pathmechdis-051122-094743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Oxygen (O2) is essential for cellular metabolism and biochemical reactions. When the demand for O2 exceeds the supply, hypoxia occurs. Hypoxia-inducible factors (HIFs) are essential to activate adaptive and survival responses following hypoxic stress. In the gut (intestines) and liver, the presence of oxygen gradients or physiologic hypoxia is necessary to maintain normal homeostasis. While physiologic hypoxia is beneficial and aids in normal functions, pathological hypoxia is harmful as it exacerbates inflammatory responses and tissue dysfunction and is a hallmark of many cancers. In this review, we discuss the role of gut and liver hypoxia-induced signaling, primarily focusing on HIFs, in the physiology and pathobiology of gut and liver diseases. Additionally, we examine the function of HIFs in various cell types during gut and liver diseases, beyond intestinal epithelial and hepatocyte HIFs. This review highlights the importance of understanding hypoxia-induced signaling in the pathogenesis of gut and liver diseases and emphasizes the potential of HIFs as therapeutic targets.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Lin X, Dai C, Chen Z, Zhang T, Pu X. Preliminary screening of biomarkers in HAPE based on quasi-targeted metabolomics. Front Physiol 2023; 14:1122026. [PMID: 36969595 PMCID: PMC10034721 DOI: 10.3389/fphys.2023.1122026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
High altitude pulmonary edema (HAPE) is a serious threat to the physical and mental health of people who quickly enter high plateaus, deserves more attention and in-depth research. In our study, through the detection of various physiological indexes and other phenotypes in a HAPE rat model, the HAPE group showed a significant decrease in oxygen partial pressure and oxygen saturation, and a significant increase in pulmonary artery pressure and lung tissue water content. The lung histomorphology showed characteristics such as pulmonary interstitial thickening and inflammatory cell infiltration. We applied quasi-targeted metabolomics to compare and analyze the components of metabolites in arterial–veinous blood in control rats and HAPE rats. Using kyoto Encyclopedia of Genes Genomes (KEGG) enrichment analysis and two machine algorithms, we speculate that after hypoxic stress and comparing arterial blood and venous blood products in rats, the metabolites were richer, indicating that normal physiological activities, such as metabolism and pulmonary circulationhad a greater impact after hypoxic stress; D-mannoseDOWN, oxidized glutathioneDOWN, glutathione disulfideDOWN, and dehydrocholic acidDOWN in arterial blood play key roles in predicting the occurrence of HAPE; in venous blood, L-leucineDOWN, L-thyroxineDOWN, and cis-4-hydroxy- D-prolineDOWN may have key roles, which can be considered biomarkers of HAPE. This result provides a new perspective for the further diagnosis and treatment of plateau disease and lays a strong foundation for further research.
Collapse
Affiliation(s)
- Xue Lin
- Department of Basic Medicine, Medical College of Qinghai University, Xining, Qinghai Province, China
- West China Hospital, Sichuan University, Chengdu, Sichuan Provience, China
| | - Chongyang Dai
- Department of Basic Medicine, Medical College of Qinghai University, Xining, Qinghai Province, China
| | - Zhi Chen
- College of Life Science, Qinghai Normal University, Xining, Qinghai Province, China
| | - Tongzuo Zhang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai Province, China
- *Correspondence: Tongzuo Zhang, ; Xiaoyan Pu,
| | - Xiaoyan Pu
- Department of Basic Medicine, Medical College of Qinghai University, Xining, Qinghai Province, China
- *Correspondence: Tongzuo Zhang, ; Xiaoyan Pu,
| |
Collapse
|
6
|
So J, Taleb S, Wann J, Strobel O, Kim K, Roh HC. Chronic cAMP activation induces adipocyte browning through discordant biphasic remodeling of transcriptome and chromatin accessibility. Mol Metab 2022; 66:101619. [PMID: 36273781 PMCID: PMC9636484 DOI: 10.1016/j.molmet.2022.101619] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Adipose tissue thermogenesis has been suggested as a new therapeutic target to promote energy metabolism for obesity and metabolic disease. Cold-inducible thermogenic adipocytes, called beige adipocytes, have attracted significant attention for their potent anti-obesity activity in adult humans. In this study, we identified the mechanisms underlying beige adipocyte recruitment, so-called adipocyte browning, by different stimuli. METHODS We generated a new adipocyte cell line with enhanced browning potentials and determined its transcriptomic and epigenomic responses following cAMP (forskolin, FSK) versus PPARγ activation (rosiglitazone). We performed time-course RNA-seq and compared the treatments and in vivo adipocyte browning. We also developed an improved protocol for Assay for Transposase Accessible Chromatin-sequencing (ATAC-seq) and defined changes in chromatin accessibility in a time course. The RNA-seq and ATAC-seq data were integrated to determine the kinetics of their coordinated regulation and to identify a transcription factor that drives these processes. We conducted functional studies using pharmacological and genetic approaches with specific inhibitors and shRNA-mediated knockdown, respectively. RESULTS FSK, not rosiglitazone, resulted in a biphasic transcriptomic response, resembling the kinetics of in vivo cold-induced browning. FSK promoted tissue remodeling first and subsequently shifted energy metabolism, concluding with a transcriptomic profile similar to that induced by rosiglitazone. The thermogenic effects of FSK were abolished by PPARγ antagonists, indicating PPARγ as a converging point. ATAC-seq uncovered that FSK leads to a significant chromatin remodeling that precedes or persists beyond transcriptomic changes, whereas rosiglitazone induces minimal changes. Motif analysis identified nuclear factor, interleukin 3 regulated (NFIL3) as a transcriptional regulator connecting the biphasic response of FSK-induced browning, as indicated by disrupted thermogenesis with NFIL3 knockdown. CONCLUSIONS Our findings elucidated unique dynamics of the transcriptomic and epigenomic remodeling in adipocyte browning, providing new mechanistic insights into adipose thermogenesis and molecular targets for obesity treatment.
Collapse
|
7
|
Wang X, Hu J, Fang Y, Fu Y, Liu B, Zhang C, Feng S, Lu X. Multi-Omics Profiling to Assess Signaling Changes upon VHL Restoration and Identify Putative VHL Substrates in Clear Cell Renal Cell Carcinoma Cell Lines. Cells 2022; 11:cells11030472. [PMID: 35159281 PMCID: PMC8833913 DOI: 10.3390/cells11030472] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/21/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
The inactivation of von Hippel–Lindau (VHL) is critical for clear cell renal cell carcinoma (ccRCC) and VHL syndrome. VHL loss leads to the stabilization of hypoxia-inducible factor α (HIFα) and other substrate proteins, which, together, drive various tumor-promoting pathways. There is inadequate molecular characterization of VHL restoration in VHL-defective ccRCC cells. The identities of HIF-independent VHL substrates remain elusive. We reinstalled VHL expression in 786-O and performed transcriptome, proteome and ubiquitome profiling to assess the molecular impact. The transcriptome and proteome analysis revealed that VHL restoration caused the downregulation of hypoxia signaling, glycolysis, E2F targets, and mTORC1 signaling, and the upregulation of fatty acid metabolism. Proteome and ubiquitome co-analysis, together with the ccRCC CPTAC data, enlisted 57 proteins that were ubiquitinated and downregulated by VHL restoration and upregulated in human ccRCC. Among them, we confirmed the reduction of TGFBI (ubiquitinated at K676) and NFKB2 (ubiquitinated at K72 and K741) by VHL re-expression in 786-O. Immunoprecipitation assay showed the physical interaction between VHL and NFKB2. K72 of NFKB2 affected NFKB2 stability in a VHL-dependent manner. Taken together, our study generates a comprehensive molecular catalog of a VHL-restored 786-O model and provides a list of putative VHL-dependent ubiquitination substrates, including TGFBI and NFKB2, for future investigation.
Collapse
Affiliation(s)
- Xuechun Wang
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (X.W.); (Y.F.)
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Jin Hu
- Mass Spectrometry & Metabolomics Core Facility, Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou 310024, China;
| | - Yihao Fang
- Department of the Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA;
| | - Yanbin Fu
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (X.W.); (Y.F.)
| | - Bing Liu
- Department of Urology, Eastern Hepatobiliary Surgery Hospital, Shanghai 201805, China;
| | - Chao Zhang
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; (X.W.); (Y.F.)
- Correspondence: (C.Z.); (S.F.); (X.L.)
| | - Shan Feng
- Mass Spectrometry & Metabolomics Core Facility, Key Laboratory of Structural Biology of Zhejiang Province, Westlake University, Hangzhou 310024, China;
- Correspondence: (C.Z.); (S.F.); (X.L.)
| | - Xin Lu
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, USA
- Correspondence: (C.Z.); (S.F.); (X.L.)
| |
Collapse
|
8
|
Demandt JAF, van Kuijk K, Theelen TL, Marsch E, Heffron SP, Fisher EA, Carmeliet P, Biessen EAL, Sluimer JC. Whole-Body Prolyl Hydroxylase Domain (PHD) 3 Deficiency Increased Plasma Lipids and Hematocrit Without Impacting Plaque Size in Low-Density Lipoprotein Receptor Knockout Mice. Front Cell Dev Biol 2021; 9:664258. [PMID: 34055796 PMCID: PMC8160238 DOI: 10.3389/fcell.2021.664258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022] Open
Abstract
Background and aims: Atherosclerosis is an important cause of clinical cardiovascular events. Atherosclerotic plaques are hypoxic, and reoxygenation improves plaque phenotype. Central players in hypoxia are hypoxia inducible factors (HIF) and their regulators, HIF-prolyl hydroxylase (PHD) isoforms 1, 2, and 3. PHD inhibitors, targeting all three isoforms, are used to alleviate anemia in chronic kidney disease. Likewise, whole-body PHD1 and PHD2ko ameliorate hypercholesterolemia and atherogenesis. As the effect of whole-body PHD3 is unknown, we investigated the effects of germline whole-body PHD3ko on atherosclerosis. Approach and Results: To initiate hypercholesterolemia and atherosclerosis low-density lipoprotein receptor knockout (LDLrko) and PHD3/LDLr double knockout (PHD3dko), mice were fed a high-cholesterol diet. Atherosclerosis and hypoxia marker pimonidazole were analyzed in aortic roots and brachiocephalic arteries. In contrast to earlier reports on PHD1- and PHD2-deficient mice, a small elevation in the body weight and an increase in the plasma cholesterol and triglyceride levels were observed after 10 weeks of diet. Dyslipidemia might be explained by an increase in hepatic mRNA expression of Cyp7a1 and fatty acid synthase, while lipid efflux of PHD3dko macrophages was comparable to controls. Despite dyslipidemia, plaque size, hypoxia, and phenotype were not altered in the aortic root or in the brachiocephalic artery of PHD3dko mice. Additionally, PHD3dko mice showed enhanced blood hematocrit levels, but no changes in circulating, splenic or lymphoid immune cell subsets. Conclusion: Here, we report that whole-body PHD3dko instigated an unfavorable lipid profile and increased hematocrit, in contrast to other PHD isoforms, yet without altering atherosclerotic plaque development.
Collapse
Affiliation(s)
- Jasper A. F. Demandt
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Kim van Kuijk
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, Netherlands
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Thomas L. Theelen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Elke Marsch
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, Netherlands
| | - Sean P. Heffron
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, Grossman School of Medicine, New York University, New York, NY, United States
| | - Edward A. Fisher
- Center for the Prevention of Cardiovascular Disease, Department of Medicine, Grossman School of Medicine, New York University, New York, NY, United States
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, Leuven, Belgium
| | - Erik A. L. Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Judith C. Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC), Maastricht, Netherlands
- BHF Centre for Cardiovascular Sciences (CVS), University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Mooli RGR, Rodriguez J, Takahashi S, Solanki S, Gonzalez FJ, Ramakrishnan SK, Shah YM. Hypoxia via ERK Signaling Inhibits Hepatic PPARα to Promote Fatty Liver. Cell Mol Gastroenterol Hepatol 2021; 12:585-597. [PMID: 33798787 PMCID: PMC8258975 DOI: 10.1016/j.jcmgh.2021.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Fatty liver or nonalcoholic fatty liver disease (NAFLD) is the most common liver disease associated with comorbidities such as insulin resistance and cardiovascular and metabolic diseases. Chronic activation of hypoxic signaling, in particular, hypoxia-inducible factor (HIF)2α, promotes NAFLD progression by repressing genes involved in fatty acid β-oxidation through unclear mechanisms. Therefore, we assessed the precise mechanism by which HIF2α promotes fatty liver and its physiological relevance in metabolic homeostasis. METHODS Primary hepatocytes from VHL (VhlΔHep) and PPARα (Ppara-null) knockout mice that were loaded with fatty acids, murine dietary protocols to induce hepatic steatosis, and fasting-refeeding dietary regimen approaches were used to test our hypothesis. RESULTS Inhibiting autophagy using chloroquine did not decrease lipid contents in VhlΔHep primary hepatocytes. Inhibition of ERK using MEK inhibitor decreased lipid contents in primary hepatocytes from a genetic model of constitutive HIF activation and primary hepatocytes loaded with free fatty acids. Moreover, MEK-ERK inhibition potentiated ligand-dependent activation of PPARα. We also show that MEK-ERK inhibition improved diet-induced hepatic steatosis, which is associated with the induction of PPARα target genes. During fasting, fatty acid β-oxidation is induced by PPARα, and refeeding inhibits β-oxidation. Our data show that ERK is involved in the post-prandial repression of hepatic PPARα signaling. CONCLUSIONS Overall, our results demonstrate that ERK activated by hypoxia signaling plays a crucial role in fatty acid β-oxidation genes by repressing hepatocyte PPARα signaling.
Collapse
Affiliation(s)
- Raja Gopal Reddy Mooli
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jessica Rodriguez
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Shogo Takahashi
- Departments of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, District of Columbia; National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sumeet Solanki
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Frank J Gonzalez
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sadeesh K Ramakrishnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
10
|
Zhao Z, Yin L, Wu F, Tong X. Hepatic metabolic regulation by nuclear factor E4BP4. J Mol Endocrinol 2021; 66:R15-R21. [PMID: 33434146 PMCID: PMC7808567 DOI: 10.1530/jme-20-0239] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
Discovered as a b-ZIP transcription repressor 30 years ago, E4 promoter-binding protein 4 (E4BP4) has been shown to play critical roles in immunity, circadian rhythms, and cancer progression. Recent research has highlighted E4BP4 as a novel regulator of metabolisms in various tissues. In this review, we focus on the function and mechanisms of hepatic E4BP4 in regulating lipid and glucose homeostasis, bile metabolism, as well as xenobiotic metabolism. Finally, E4BP4-specific targets will be discussed for the prevention and treatment of metabolic disorders.
Collapse
Affiliation(s)
- Zifeng Zhao
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, P. R. China 211198
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Lei Yin
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Feihua Wu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, P. R. China 211198
| | - Xin Tong
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
11
|
Baik AH, Jain IH. Turning the Oxygen Dial: Balancing the Highs and Lows. Trends Cell Biol 2020; 30:516-536. [PMID: 32386878 PMCID: PMC7391449 DOI: 10.1016/j.tcb.2020.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/02/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Oxygen is both vital and toxic to life. Molecular oxygen is the most used substrate in the human body and is required for several hundred diverse biochemical reactions. The discovery of the PHD-HIF-pVHL system revolutionized our fundamental understanding of oxygen sensing and cellular adaptations to hypoxia. It deepened our knowledge of the biochemical underpinnings of numerous diseases, ranging from anemia to cancer. Cellular dysfunction and tissue pathology can result from a mismatch of oxygen supply and demand. Recent work has shown that mitochondrial disease models display tissue hyperoxia and that disease pathology can be reversed by normalization of excess oxygen, suggesting that certain disease states can potentially be treated by modulating oxygen levels. In this review, we describe cellular and organismal mechanisms of oxygen sensing and adaptation. We provide a revitalized framework for understanding pathologies of too little or too much oxygen.
Collapse
Affiliation(s)
- Alan H Baik
- Department of Physiology, University of California, San Francisco, CA 94158, USA; Department of Medicine, Division of Cardiology, University of California, San Francisco, CA 94143, USA.
| | - Isha H Jain
- Department of Physiology, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
12
|
Li Y, Chen Y, Huang X, Huang D, Gan H, Yao N, Hu Z, Li R, Zhan X, Xie K, Jiang J, Cai D. Tanshinol A Ameliorates Triton-1339W-Induced Hyperlipidemia and Liver Injury in C57BL/6J Mice by Regulating mRNA Expression of Lipemic-Oxidative Injury Genes. Lipids 2020; 55:127-140. [PMID: 32058595 DOI: 10.1002/lipd.12217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 12/14/2022]
Abstract
Tanshinol A, which is derived from a traditional Chinese herbal Radix Salviae Miltiorrhizae is indicative of a hypolipidemic candidate. Therefore, we aim to validate its hypolipidemic activity of tanshinol A and explore its mechanism in triton-1339W-induced hyperlipidemic mice model, which possess multiply pathogenesis for endogenous lipid metabolism disorder. Experimental hyperlipidemia mice are treated with or without tanshinol A (i.g. 40, 20, 10 mg/kg), and blood and liver tissue were collected for validating its hypolipidemic and hepatic protective effect, and hepatic mRNA expression profile, which was associated with lipid metabolism dysfunction and liver injury, was detected by RT-qPCR. As results show, triton-1339W-induced abnormal of serum TC, TAG, HDL-C, LDL-C, SOD, MDA, GOT, and GPT is remarkably attenuated by tanshinol A. In pathological experiment, triton-1339W-induced hepatocellular ballooning degeneration, irregular central vein congestion, and inflammation infiltration are alleviated by tanshinol A. Correspondingly, hepatic mRNA expression of Atf4, Fgf21, Vldlr, Nqo1, Pdk4, and Angptl4, which are genes regulating lipemic-oxidative injury, are significantly increased by tanshinol A by 2~6 fold. Abcg5, Cd36, and Apob, which are responsible for cholesterol metabolism, are mildly upregulated. Noticeably, triton-1339W-suppressed expressions of Ptgs2/Il10, which are genes responsible for acute inflammation resolution in liver injury, are remarkably increased by tanshinol A. Conclusively, tanshinol A exerted hypolipidemic effect and hepatoprotective effect through restoring triton-1339W-suppressed mRNA expression, which may be involved in Atf4/Fgf21/Vldlr and Ptgs2/Il-10 signaling pathways.
Collapse
Affiliation(s)
- Yuting Li
- Department of Pharmacology of Traditional Chinese Medicine, The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Yuxing Chen
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Xuejun Huang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Dane Huang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Haining Gan
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Nan Yao
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Zixuan Hu
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Ruyue Li
- Department of Pharmacology of Traditional Chinese Medicine, The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Xinyi Zhan
- Department of Pharmacology of Traditional Chinese Medicine, The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Kaifeng Xie
- Department of Pharmacology of Traditional Chinese Medicine, The Fifth Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Jieyi Jiang
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| | - Dake Cai
- Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China.,Guangdong Provincial key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, 510095, China
| |
Collapse
|
13
|
Zuniga-Hertz JP, Patel HH. The Evolution of Cholesterol-Rich Membrane in Oxygen Adaption: The Respiratory System as a Model. Front Physiol 2019; 10:1340. [PMID: 31736773 PMCID: PMC6828933 DOI: 10.3389/fphys.2019.01340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022] Open
Abstract
The increase in atmospheric oxygen levels imposed significant environmental pressure on primitive organisms concerning intracellular oxygen concentration management. Evidence suggests the rise of cholesterol, a key molecule for cellular membrane organization, as a cellular strategy to restrain free oxygen diffusion under the new environmental conditions. During evolution and the increase in organismal complexity, cholesterol played a pivotal role in the establishment of novel and more complex functions associated with lipid membranes. Of these, caveolae, cholesterol-rich membrane domains, are signaling hubs that regulate important in situ functions. Evolution resulted in complex respiratory systems and molecular response mechanisms that ensure responses to critical events such as hypoxia facilitated oxygen diffusion and transport in complex organisms. Caveolae have been structurally and functionally associated with respiratory systems and oxygen diffusion control through their relationship with molecular response systems like hypoxia-inducible factors (HIF), and particularly as a membrane-localized oxygen sensor, controlling oxygen diffusion balanced with cellular physiological requirements. This review will focus on membrane adaptations that contribute to regulating oxygen in living systems.
Collapse
Affiliation(s)
- Juan Pablo Zuniga-Hertz
- Department of Anesthesiology, VA San Diego Healthcare System, University of California, San Diego, San Diego, CA, United States
| | - Hemal H Patel
- Department of Anesthesiology, VA San Diego Healthcare System, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
14
|
Seim GL, Britt EC, John SV, Yeo FJ, Johnson AR, Eisenstein RS, Pagliarini DJ, Fan J. Two-stage metabolic remodelling in macrophages in response to lipopolysaccharide and interferon-γ stimulation. Nat Metab 2019; 1:731-742. [PMID: 32259027 PMCID: PMC7108803 DOI: 10.1038/s42255-019-0083-2] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 06/07/2019] [Indexed: 12/13/2022]
Abstract
In response to signals associated with infection or tissue damage, macrophages undergo a series of dynamic phenotypic changes. Here we show that during the response to LPS and interferon-γ stimulation, metabolic reprogramming in macrophages is also highly dynamic. Specifically, the TCA cycle undergoes a two-stage remodeling: the early stage is characterized by a transient accumulation of intermediates including succinate and itaconate, while the late stage is marked by the subsidence of these metabolites. The metabolic transition into the late stage is largely driven by the inhibition of pyruvate dehydrogenase complex (PDHC) and oxoglutarate dehydrogenase complex (OGDC), which is controlled by the dynamic changes in lipoylation state of both PDHC and OGDC E2 subunits and phosphorylation of PDHC E1 subunit. This dynamic metabolic reprogramming results in a transient metabolic state that strongly favors HIF-1α stabilization during the early stage, which subsides by the late stage; consistently, HIF-1α levels follow this trend. This study elucidates a dynamic and mechanistic picture of metabolic reprogramming in LPS and interferon-γ stimulated macrophages, and provides insights into how changing metabolism can regulate the functional transitions in macrophages over a course of immune response.
Collapse
Affiliation(s)
- Gretchen L Seim
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Emily C Britt
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Steven V John
- Morgridge Institute for Research, Madison, WI
- Cell and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI
| | | | | | - Richard S Eisenstein
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- Morgridge Institute for Research, Madison, WI
| | - David J Pagliarini
- Morgridge Institute for Research, Madison, WI
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, Madison, WI
- Correspondence to
| |
Collapse
|
15
|
Wu N, McDaniel K, Zhou T, Ramos-Lorenzo S, Wu C, Huang L, Chen D, Annable T, Francis H, Glaser S, Alpini G, Meng F. Knockout of microRNA-21 attenuates alcoholic hepatitis through the VHL/NF-κB signaling pathway in hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol 2018; 315:G385-G398. [PMID: 29848019 PMCID: PMC6415712 DOI: 10.1152/ajpgi.00111.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/21/2018] [Accepted: 05/27/2018] [Indexed: 01/31/2023]
Abstract
microRNA-21 (miRNA) is one of the most abundant miRNAs in chronic liver injuries including alcoholic liver injury. Previous studies have demonstrated that miR-21 plays a role in inflammation in the liver and functions in hepatic stellate cells (HSCs), which reside in the perisinusoidal space between sinusoidal endothelial cells and hepatocytes and regulate sinusoidal circulation. HSCs integrate cytokine-mediated inflammatory responses in the sinusoids and relay them to the liver parenchyma. Here, we showed that the activation of Von Hippel-Lindau (VHL) expression, by miR-21 knockout in vivo and anti-miR-21 or VHL overexpression in vitro, suppressed the production of proinflammatory cytokines, such as interleukin (IL)-6, monocyte chemoattractant protein-1, and IL-1β, in human HSCs during alcoholic liver injury. Sequence and functional analyses confirmed that miR-21 directly targeted the 3'-untranslated region of VHL. Immunofluorescence and real-time PCR analysis revealed that miR-21 depletion blocked NF-κB activation in human HSCs both in cultured HSCs as well as HSCs isolated from alcohol-related liver disease mice liver by laser capture microdissection. We also showed that conditioned medium from anti-miR-21-transfected HSCs suppressed human monocyte-derived THP-1 cell migration. Taken together, our study indicates that depletion of miR-21 may downregulate cytokine production in HSCs and macrophage chemotaxis during alcoholic liver injury and that the targeting of miR-21 may have therapeutic potential for preventing the progression of alcoholic liver diseases. NEW & NOTEWORTHY This study demonstrates that silencing microRNA-21 can inhibit cytokine production and inflammatory responses in human hepatic stellate cells during alcoholic liver injury and that the targeting of microR-21 in hepatic stellate cells may have therapeutic potential for prevention and treatment of alcoholic liver diseases.
Collapse
Affiliation(s)
- Nan Wu
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
| | - Kelly McDaniel
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
- Research Institute, Baylor Scott & White Health, Temple, Texas
| | - Tianhao Zhou
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
| | - Sugeily Ramos-Lorenzo
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
- Research Institute, Baylor Scott & White Health, Temple, Texas
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University , College Station, Texas
| | - Li Huang
- Department of Hepatobiliary Surgery and Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University , Guangdong , China
| | - Demeng Chen
- Department of Hepatobiliary Surgery and Center for Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University , Guangdong , China
| | - Tami Annable
- Research Institute, Baylor Scott & White Health, Temple, Texas
- Texas Bioscience District, Temple, Texas
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
- Research Institute, Baylor Scott & White Health, Temple, Texas
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Department of Medicine and Baylor Scott & White Digestive Disease Research Center, Texas A&M Health Sciences Center and Scott & White Hospital, Temple, Texas
- Research Institute, Baylor Scott & White Health, Temple, Texas
| |
Collapse
|
16
|
Xie C, Gao X, Sun D, Zhang Y, Krausz KW, Qin X, Gonzalez FJ. Metabolic Profiling of the Novel Hypoxia-Inducible Factor 2 α Inhibitor PT2385 In Vivo and In Vitro. Drug Metab Dispos 2018; 46:336-345. [PMID: 29363499 DOI: 10.1124/dmd.117.079723] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/18/2018] [Indexed: 12/13/2022] Open
Abstract
PT2385 is a first-in-class, selective small-molecule inhibitor of hypoxia-inducible factor-2α (HIF-2α) developed for the treatment of advanced clear cell renal cell carcinoma. Preclinical results demonstrated that PT2385 has potent antitumor efficacy in mouse xenograft models of kidney cancer. It also has activity toward metabolic disease in a mouse model. However, no metabolism data are currently publically available. It is of great importance to characterize the metabolism of PT2385 and identify its effect on systemic homeostasis in mice. High-resolution mass spectrometry-based metabolomics was performed to profile the biotransformation of PT2385 and PT2385-induced changes in endogenous metabolites. Liver microsomes and recombinant drug-metabolizing enzymes were used to determine the mechanism of PT2385 metabolism. Real-time polymerase chain reaction analysis was employed to investigate the reason for the PT2385-induced bile acid dysregulation. A total of 12 metabolites of PT2385 was characterized, generated from hydroxylation (M1, M2), dihydroxylation and desaturation (M3, M4), oxidative-defluorination (M7), glucuronidation (M8), N-acetylcysteine conjugation (M9), and secondary methylation (M5, M6) and glucuronidation (M10, M11, and M12). CYP2C19 was the major contributor to the formation of M1, M2, and M7, UGT2B17 to M8, and UGT1A1/3 to M10-M12. The bile acid metabolites taurocholic acid and tauro-β-muricholic acid were elevated in serum and liver of mice after PT2385 treatment. Gene expression analysis further revealed that intestinal HIF-2α inhibition by PT2385 treatment upregulated the hepatic expression of CYP7A1, the rate-limiting enzyme in bile acid synthesis. This study provides metabolic data and an important reference basis for the safety evaluation and rational clinical application of PT2385.
Collapse
Affiliation(s)
- Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| | - Xiaoxia Gao
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| | - Dongxue Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| | - Youbo Zhang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| | - Xuemei Qin
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (C.X., X.G., D.S., Y.Z., K.W.K., F.J.G.); and Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, Shanxi, People's Republic of China (X.G., X.Q.)
| |
Collapse
|
17
|
Ramakrishnan SK, Shah YM. A central role for hypoxia-inducible factor (HIF)-2α in hepatic glucose homeostasis. ACTA ACUST UNITED AC 2017; 4:207-216. [PMID: 29276790 PMCID: PMC5734117 DOI: 10.3233/nha-170022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hepatic glucose production is regulated by hormonal and dietary factors. At fasting, 80% of glucose released into the circulation is derived from the liver, among which gluconeogenesis accounts for 55% and the rest by glycogenolysis. Studies suggest a complex mechanism involved in the regulation of hepatic glucose metabolism during fasting and post-absorptive phase. Oxygen plays a key role in numerous metabolic pathways such as TCA cycle, gluconeogenesis, glycolysis and fatty acid oxidation. Oxygenation of the gastrointestinal tract including liver and intestine is dynamically regulated by changes in the blood flow and metabolic activity. Cellular adaptation to low oxygen is mediated by the transcription factors HIF-1α and HIF-2α. HIF-1α regulates glycolytic genes whereas HIF-2α is known to primarily regulate genes involved in cell proliferation and iron metabolism. This review focuses on the role of the oxygen sensing signaling in the regulation of hepatic glucose output with an emphasis on hypoxia inducible factor (HIF)-2α. Recent studies have established a metabolic role of HIF-2α in systemic glucose homeostasis. Understanding the HIF-2α dependent mechanism in hepatic metabolism will greatly enhance our potential to utilize the oxygen sensing mechanisms to treat metabolic diseases.
Collapse
Affiliation(s)
- Sadeesh K Ramakrishnan
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.,Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
18
|
Martin ER, Smith MT, Maroni BJ, Zuraw QC, deGoma EM. Clinical Trial of Vadadustat in Patients with Anemia Secondary to Stage 3 or 4 Chronic Kidney Disease. Am J Nephrol 2017; 45:380-388. [PMID: 28343225 DOI: 10.1159/000464476] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Therapeutic options for the treatment of anemia secondary to chronic kidney disease (CKD) remain limited. Vadadustat (AKB-6548) is an oral hypoxia-inducible factor prolyl-hydroxylase domain (HIF-PHD) inhibitor that is being investigated for the treatment of anemia secondary to CKD. METHODS A phase 2a, multicenter, randomized, double-blind, placebo-controlled, dose-ranging trial (NCT01381094) was undertaken in adults with anemia secondary to CKD stage 3 or 4. Eligible subjects were evenly randomized to 5 groups: 240, 370, 500, or 630 mg of once-daily oral vadadustat or placebo for 6 weeks. All subjects received low-dose supplemental oral iron (50 mg daily). The primary endpoint was the mean absolute change in hemoglobin (Hb) from baseline to the end of treatment. Secondary endpoints included iron indices, safety, and tolerability. RESULTS Ninety-three subjects were randomized. Compared with placebo, vadadustat significantly increased Hb after 6 weeks in a dose-dependent manner (analysis of variance; p < 0.0001). Vadadustat increased the total iron-binding capacity and decreased concentrations of ferritin and hepcidin. The proportion of subjects with at least 1 treatment-emergent adverse event was similar between vadadustat- and placebo-treated groups. No significant changes in blood pressure, vascular endothelial growth factor, C-reactive protein, or total cholesterol were observed. Limitations of this study included its small sample size and short treatment duration. CONCLUSIONS Vadadustat increased Hb levels and improved biomarkers of iron mobilization and utilization in patients with anemia secondary to stage 3 or 4 CKD. Global multicenter, randomized phase 3 trials are ongoing in non-dialysis-dependent and dialysis-dependent patients.
Collapse
|
19
|
Moon Y, Park B, Park H. Hypoxic repression of CYP7A1 through a HIF-1α- and SHP-independent mechanism. BMB Rep 2017; 49:173-8. [PMID: 26521940 PMCID: PMC4915232 DOI: 10.5483/bmbrep.2016.49.3.188] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Indexed: 01/05/2023] Open
Abstract
Liver cells experience hypoxic stress when drug-metabolizing enzymes excessively consume O2 for hydroxylation. Hypoxic stress changes the transcription of several genes by activating a heterodimeric transcription factor called hypoxia-inducible factor- 1α/β (HIF-1α/β). We found that hypoxic stress (0.1% O2) decreased the expression of cytochrome P450 7A1 (CYP7A1), a rate-limiting enzyme involved in bile acid biosynthesis. Chenodeoxycholic acid (CDCA), a major component of bile acids, represses CYP7A1 by activating a transcriptional repressor named small heterodimer partner (SHP). We observed that hypoxia decreased the levels of both CDCA and SHP, suggesting that hypoxia repressed CYP7A1 without inducing SHP. The finding that overexpression of HIF-1α increased the activity of the CYP7A1 promoter suggested that hypoxia decreased the expression of CYP7A1 in a HIF-1-independent manner. Thus, the results of this study suggested that hypoxia decreased the activity of CYP7A1 by limiting its substrate O2, and by decreasing the transcription of CYP7A1. [BMB Reports 2016; 49(3): 173-178].
Collapse
Affiliation(s)
- Yunwon Moon
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Bongju Park
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 02504, Korea
| |
Collapse
|
20
|
Rahtu-Korpela L, Karsikas S, Hörkkö S, Blanco Sequeiros R, Lammentausta E, Mäkelä KA, Herzig KH, Walkinshaw G, Kivirikko KI, Myllyharju J, Serpi R, Koivunen P. HIF prolyl 4-hydroxylase-2 inhibition improves glucose and lipid metabolism and protects against obesity and metabolic dysfunction. Diabetes 2014; 63:3324-33. [PMID: 24789921 DOI: 10.2337/db14-0472] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Obesity is a major public health problem, predisposing subjects to metabolic syndrome, type 2 diabetes, and cardiovascular diseases. Specific prolyl 4-hydroxylases (P4Hs) regulate the stability of the hypoxia-inducible factor (HIF), a potent governor of metabolism, with isoenzyme 2 being the main regulator. We investigated whether HIF-P4H-2 inhibition could be used to treat obesity and its consequences. Hif-p4h-2-deficient mice, whether fed normal chow or a high-fat diet, had less adipose tissue, smaller adipocytes, and less adipose tissue inflammation than their littermates. They also had improved glucose tolerance and insulin sensitivity. Furthermore, the mRNA levels of the HIF-1 targets glucose transporters, glycolytic enzymes, and pyruvate dehydrogenase kinase-1 were increased in their tissues, whereas acetyl-CoA concentration was decreased. The hepatic mRNA level of the HIF-2 target insulin receptor substrate-2 was higher, whereas that of two key enzymes of fatty acid synthesis was lower. Serum cholesterol levels and de novo lipid synthesis were decreased, and the mice were protected against hepatic steatosis. Oral administration of an HIF-P4H inhibitor, FG-4497, to wild-type mice with metabolic dysfunction phenocopied these beneficial effects. HIF-P4H-2 inhibition may be a novel therapy that not only protects against the development of obesity and its consequences but also reverses these conditions.
Collapse
Affiliation(s)
- Lea Rahtu-Korpela
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| | - Sara Karsikas
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| | - Sohvi Hörkkö
- Nordlab Oulu, Oulu University Hospital, FIN-90220 Oulu, Finland Department of Medical Microbiology and Immunology, Medical Research Center, University of Oulu, FIN-90014 Oulu, Finland
| | - Roberto Blanco Sequeiros
- Department of Radiology, Oulu University Hospital and University of Oulu, FIN-90029 Oulu, Finland
| | - Eveliina Lammentausta
- Department of Radiology, Oulu University Hospital and University of Oulu, FIN-90029 Oulu, Finland
| | - Kari A Mäkelä
- Biocenter Oulu, Department of Physiology, University of Oulu, FIN-90014 Oulu, Finland
| | - Karl-Heinz Herzig
- Biocenter Oulu, Department of Physiology, University of Oulu, FIN-90014 Oulu, Finland
| | | | - Kari I Kivirikko
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| | - Johanna Myllyharju
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| | - Raisa Serpi
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| | - Peppi Koivunen
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, FIN-90014 Oulu, Finland
| |
Collapse
|
21
|
Hypoxia-inducible factor 2 alpha is essential for hepatic outgrowth and functions via the regulation of leg1 transcription in the zebrafish embryo. PLoS One 2014; 9:e101980. [PMID: 25000307 PMCID: PMC4084947 DOI: 10.1371/journal.pone.0101980] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 06/13/2014] [Indexed: 12/11/2022] Open
Abstract
The liver plays a vital role in metabolism, detoxification, digestion, and the maintenance of homeostasis. During development, the vertebrate embryonic liver undergoes a series of morphogenic processes known as hepatogenesis. Hepatogenesis can be separated into three interrelated processes: endoderm specification, hepatoblast differentiation, and hepatic outgrowth. Throughout this process, signaling molecules and transcription factors initiate and regulate the coordination of cell proliferation, apoptosis, differentiation, intercellular adhesion, and cell migration. Hifs are already recognized to be essential in embryonic development, but their role in hepatogenesis remains unknown. Using the zebrafish embryo as a model organism, we report that the lack of Hif2-alpha but not Hif1-alpha blocks hepatic outgrowth. While Hif2-alpha is not involved in hepatoblast specification, this transcription factor regulates hepatocyte cell proliferation during hepatic outgrowth. Furthermore, we demonstrated that the lack of Hif2-alpha can reduce the expression of liver-enriched gene 1 (leg1), which encodes a secretory protein essential for hepatic outgrowth. Additionally, exogenous mRNA expression of leg1 can rescue the small liver phenotype of hif2-alpha morphants. We also showed that Hif2-alpha directly binds to the promoter region of leg1 to control leg1 expression. Interestingly, we discovered overrepresented, high-density Hif-binding sites in the potential upstream regulatory sequences of leg1 in teleosts but not in terrestrial mammals. We concluded that hif2-alpha is a key factor required for hepatic outgrowth and regulates leg1 expression in zebrafish embryos. We also proposed that the hif2-alpha-leg1 axis in liver development may have resulted from the adaptation of teleosts to their environment.
Collapse
|
22
|
Hypoxia-inducible factor/MAZ-dependent induction of caveolin-1 regulates colon permeability through suppression of occludin, leading to hypoxia-induced inflammation. Mol Cell Biol 2014; 34:3013-23. [PMID: 24891620 DOI: 10.1128/mcb.00324-14] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Caveolae are specialized microdomains on membranes that are critical for signal transduction, cholesterol transport, and endocytosis. Caveolin-1 (CAV1) is a multifunctional protein and a major component of caveolae. Cav1 is directly activated by hypoxia-inducible factor (HIF). HIFs are heterodimers of an oxygen-sensitive α subunit, HIF1α or HIF2α, and a constitutively expressed β subunit, aryl hydrocarbon receptor nuclear translocator (ARNT). Whole-genome expression analysis demonstrated that Cav1 is highly induced in mouse models of constitutively activated HIF signaling in the intestine. Interestingly, Cav1 was increased only in the colon and not in the small intestine. Currently, the mechanism and role of HIF induction of CAV1 in the colon are unclear. In mouse models, mice that overexpressed HIF1α or HIF2α specifically in intestinal epithelial cells demonstrated an increase in Cav1 gene expression in the colon but not in the duodenum, jejunum, or ileum. HIF2α activated the Cav1 promoter in a HIF response element-independent manner. myc-associated zinc finger (MAZ) protein was essential for HIF2α activation of the Cav1 promoter. Hypoxic induction of CAV1 in the colon was essential for intestinal barrier integrity by regulating occludin expression. This may provide an additional mechanism by which chronic hypoxia can activate intestinal inflammation.
Collapse
|