1
|
Chan P, Cheung PHH, Kang XZ, Cheng Y, Wong CM, Jin DY, Chan CP. Cgref1 is a CREB-H-regulated hepatokine that promotes hepatic de novo lipogenesis by mediating epididymal fat insulin resistance. Int J Biol Sci 2025; 21:2568-2588. [PMID: 40303310 PMCID: PMC12035884 DOI: 10.7150/ijbs.97008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
Rationale: Type 2 diabetes mellitus and metabolic dysfunction-associated steatotic liver disease (MASLD) are interrelated metabolic disorders that pose significant health concerns. Hepatokines and other regulatory factors implicated in these diseases are incompletely understood. Here, we report on a new hepatokine named cell growth regulator with EF-hand domain 1 (Cgref1) that modulates lipid metabolism to aggravate these conditions. Methods: Cgref1 was identified by microarray analysis of downregulated genes in liver of Creb3l3 -/- mice. Cgref1 -/- mice were subjected to transcriptomic, metabolomic and lipidomic analyses as well as metabolic assays. Gain-of-function and loss-of-function assays were performed in primary hepatocytes and cultured human and mouse cells. Results: Cgref1 expression is induced by hepatic transcription factor CREB-H. Secreted Cgref1 primarily targets epididymal white adipose tissue (eWAT), where insulin signalling and glucose uptake are suppressed. Cgref1-/- mice showed lower tendencies of developing obesity, hyperglycaemia and dyslipidaemia, associated with compromised hepatic de novo lipogenesis. Thus, Cgref1 poses an advantage to maintain the normal functioning of vital organs by preserving glucose from being absorbed into eWAT. However, in circumstances where Cgref1 expression becomes excessive, eWAT develops insulin resistance, which in turn promotes hepatic glucose production, lipogenesis and MASLD development. Conclusion: As a hepatokine that affects blood glucose levels and lipogenesis, Cgref1 is a potential target in the intervention of metabolic disorders.
Collapse
Affiliation(s)
- Pearl Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Pak-Hin Hinson Cheung
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Xiao-Zhuo Kang
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Yun Cheng
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Chi-Ming Wong
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, 11 Yuk Choi Road, Hung Hom, Hong Kong
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| |
Collapse
|
2
|
Kim H, Chen Q, Ju D, Purandare N, Chen X, Samavati L, Li L, Zhang R, Grossman LI, Zhang K. ER-tethered stress sensor CREBH regulates mitochondrial unfolded protein response to maintain energy homeostasis. Proc Natl Acad Sci U S A 2024; 121:e2410486121. [PMID: 39589874 PMCID: PMC11626163 DOI: 10.1073/pnas.2410486121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
The Mitochondrial Unfolded Protein Response (UPRmt), a mitochondria-originated stress response to altered mitochondrial proteostasis, plays important roles in various pathophysiological processes. In this study, we revealed that the endoplasmic reticulum (ER)-tethered stress sensor CREBH regulates UPRmt to maintain mitochondrial homeostasis and function in the liver. CREBH is enriched in and required for hepatic Mitochondria-Associated Membrane (MAM) expansion induced by energy demands. Under a fasting challenge or during the circadian cycle, CREBH is activated to promote expression of the genes encoding the key enzymes, chaperones, and regulators of UPRmt in the liver. Activated CREBH, cooperating with peroxisome proliferator-activated receptor α (PPARα), activates expression of Activating Transcription Factor (ATF) 5 and ATF4, two major UPRmt transcriptional regulators, independent of the ER-originated UPR (UPRER) pathways. Hepatic CREBH deficiency leads to accumulation of mitochondrial unfolded proteins, decreased mitochondrial membrane potential, and elevated cellular redox state. Dysregulation of mitochondrial function caused by CREBH deficiency coincides with increased hepatic mitochondrial oxidative phosphorylation (OXPHOS) but decreased glycolysis. CREBH knockout mice display defects in fatty acid oxidation and increased reliance on carbohydrate oxidation for energy production. In summary, our studies uncover that hepatic UPRmt is activated through CREBH under physiological challenges, highlighting a molecular link between ER and mitochondria in maintaining mitochondrial proteostasis and energy homeostasis under stress conditions.
Collapse
Affiliation(s)
- Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Qi Chen
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Donghong Ju
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Neeraja Purandare
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Xuequn Chen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI48201
| | - Lobelia Samavati
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Li Li
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Lawrence I. Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI48201
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI48201
| |
Collapse
|
3
|
Gao YP, Hu C, Hu M, Dong WS, Li K, Ye YJ, Hu YX, Zhang X. CREB3 protein family: the promising therapeutic targets for cardiovascular and metabolic diseases. Cell Biol Toxicol 2024; 40:103. [PMID: 39581923 PMCID: PMC11586310 DOI: 10.1007/s10565-024-09939-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/17/2024] [Indexed: 11/26/2024]
Abstract
Significant advancements in cardiovascular and metabolic disease research have been made with the CREB3 protein family. Studies have revealed that members of this family are crucial in the development of these diseases, contributing to the regulation of lipid metabolism, inflammation, and vascular function. These studies provide useful information for future therapeutic strategies in cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Yi-Peng Gao
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Can Hu
- Department of Ultrasound, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Hu
- Department of Cardiology, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen-Sheng Dong
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kang Li
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu-Xin Hu
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xin Zhang
- Department of Geriatrics, Hubei Key Laboratory of Metabolic and Chronic Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
4
|
Białek W, Hryniewicz-Jankowska A, Czechowicz P, Sławski J, Collawn JF, Czogalla A, Bartoszewski R. The lipid side of unfolded protein response. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159515. [PMID: 38844203 DOI: 10.1016/j.bbalip.2024.159515] [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: 02/28/2024] [Revised: 04/16/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Although our current knowledge of the molecular crosstalk between the ER stress, the unfolded protein response (UPR), and lipid homeostasis remains limited, there is increasing evidence that dysregulation of either protein or lipid homeostasis profoundly affects the other. Most research regarding UPR signaling in human diseases has focused on the causes and consequences of disrupted protein folding. The UPR itself consists of very complex pathways that function to not only maintain protein homeostasis, but just as importantly, modulate lipid biogenesis to allow the ER to adjust and promote cell survival. Lipid dysregulation is known to activate many aspects of the UPR, but the complexity of this crosstalk remains a major research barrier. ER lipid disequilibrium and lipotoxicity are known to be important contributors to numerous human pathologies, including insulin resistance, liver disease, cardiovascular diseases, neurodegenerative diseases, and cancer. Despite their medical significance and continuous research, however, the molecular mechanisms that modulate lipid synthesis during ER stress conditions, and their impact on cell fate decisions, remain poorly understood. Here we summarize the current view on crosstalk and connections between altered lipid metabolism, ER stress, and the UPR.
Collapse
Affiliation(s)
- Wojciech Białek
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | | | - Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Aleksander Czogalla
- Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Rafał Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland.
| |
Collapse
|
5
|
Zhang R, Zhang K. A unified model for regulating lipoprotein lipase activity. Trends Endocrinol Metab 2024; 35:490-504. [PMID: 38521668 PMCID: PMC11663433 DOI: 10.1016/j.tem.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/25/2024]
Abstract
The regulation of triglyceride (TG) tissue distribution, storage, and utilization, a fundamental process of energy homeostasis, critically depends on lipoprotein lipase (LPL). We review the intricate mechanisms by which LPL activity is regulated by angiopoietin-like proteins (ANGPTL3, 4, 8), apolipoproteins (APOA5, APOC3, APOC2), and the cAMP-responsive element-binding protein H (CREBH). ANGPTL8 functions as a molecular switch, through complex formation, activating ANGPTL3 while deactivating ANGPTL4 in their LPL inhibition. The ANGPTL3-4-8 model integrates the roles of the aforementioned proteins in TG partitioning between white adipose tissue (WAT) and oxidative tissues (heart and skeletal muscles) during the feed/fast cycle. This model offers a unified perspective on LPL regulation, providing insights into TG metabolism, metabolic diseases, and therapeutics.
Collapse
Affiliation(s)
- Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Xiang X, Ji R, Han S, Xu X, Zhu S, Li Y, Du J, Mai K, Ai Q. Differences in diacylglycerol acyltransferases expression patterns and regulation cause distinct hepatic triglyceride deposition in fish. Commun Biol 2024; 7:480. [PMID: 38641731 PMCID: PMC11031565 DOI: 10.1038/s42003-024-06022-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 03/07/2024] [Indexed: 04/21/2024] Open
Abstract
Triglyceride (TAG) deposition in the liver is associated with metabolic disorders. In lower vertebrate, the propensity to accumulate hepatic TAG varies widely among fish species. Diacylglycerol acyltransferases (DGAT1 and DGAT2) are major enzymes for TAG synthesis. Here we show that large yellow croaker (Larimichthys crocea) has significantly higher hepatic TAG level than that in rainbow trout (Oncorhynchus mykiss) fed with same diet. Hepatic expression of DGATs genes in croaker is markedly higher compared with trout under physiological condition. Meanwhile, DGAT1 and DGAT2 in both croaker and trout are required for TAG synthesis and lipid droplet formation in vitro. Furthermore, oleic acid treatment increases DGAT1 expression in croaker hepatocytes rather than in trout and has no significant difference in DGAT2 expression in two fish species. Finally, effects of various transcription factors on croaker and trout DGAT1 promoter are studied. We find that DGAT1 is a target gene of the transcription factor CREBH in croaker rather than in trout. Overall, hepatic expression and transcriptional regulation of DGATs display significant species differences between croaker and trout with distinct hepatic triglyceride deposition, which bring new perspectives on the use of fish models for studying hepatic TAG deposition.
Collapse
Affiliation(s)
- Xiaojun Xiang
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Renlei Ji
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Shangzhe Han
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Xiang Xu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Si Zhu
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Yongnan Li
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Jianlong Du
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
| | - Kangsen Mai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong, 266237, People's Republic of China
| | - Qinghui Ai
- Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture and Rural Affairs) & Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao, Shandong, 266003, P.R. China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, Shandong, 266237, People's Republic of China.
| |
Collapse
|
7
|
Sun Y, Zhang Y, Zhang J, Chen YE, Jin JP, Zhang K, Mou H, Liang X, Xu J. XBP1-mediated transcriptional regulation of SLC5A1 in human epithelial cells in disease conditions. Cell Biosci 2024; 14:27. [PMID: 38388523 PMCID: PMC10885492 DOI: 10.1186/s13578-024-01203-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Sodium-Glucose cotransporter 1 and 2 (SGLT1/2) belong to the family of glucose transporters, encoded by SLC5A1 and SLC5A2, respectively. SGLT2 is almost exclusively expressed in the renal proximal convoluted tubule cells. SGLT1 is expressed in the kidneys but also in other organs throughout the body. Many SGLT inhibitor drugs have been developed based on the mechanism of blocking glucose (re)absorption mediated by SGLT1/2, and several have gained major regulatory agencies' approval for treating diabetes. Intriguingly these drugs are also effective in treating diseases beyond diabetes, for example heart failure and chronic kidney disease. We recently discovered that SGLT1 is upregulated in the airway epithelial cells derived from patients of cystic fibrosis (CF), a devastating genetic disease affecting greater than 70,000 worldwide. RESULTS In the present work, we show that the SGLT1 upregulation is coupled with elevated endoplasmic reticulum (ER) stress response, indicated by activation of the primary ER stress senor inositol-requiring protein 1α (IRE1α) and the ER stress-induced transcription factor X-box binding protein 1 (XBP1), in CF epithelial cells, and in epithelial cells of other stress conditions. Through biochemistry experiments, we demonstrated that the spliced form of XBP1 (XBP1s) acts as a transcription factor for SLC5A1 by directly binding to its promoter region. Targeting this ER stress → SLC5A1 axis by either the ER stress inhibitor Rapamycin or the SGLT1 inhibitor Sotagliflozin was effective in attenuating the ER stress response and reducing the SGLT1 level in these cellular model systems. CONCLUSIONS The present work establishes a causal relationship between ER stress and SGLT1 upregulation and provides a mechanistic explanation why SGLT inhibitor drugs benefit diseases beyond diabetes.
Collapse
Affiliation(s)
- Yifei Sun
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yihan Zhang
- The Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Jackson, 1402, Boston, MA, 02114, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jian-Ping Jin
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Hongmei Mou
- The Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Jackson, 1402, Boston, MA, 02114, USA.
| | - Xiubin Liang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Deng X, Liu B, Jiang Q, Li G, Li J, Xu K. CREBH promotes autophagy to ameliorate NASH by regulating Coro1a. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166914. [PMID: 37837948 DOI: 10.1016/j.bbadis.2023.166914] [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: 03/13/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Dysfunctional autophagy aggravates oxidative stress and inflammation in hepatocytes and accelerates the progression of nonalcoholic steatohepatitis (NASH). Here, we demonstrated that cAMP-responsive element-binding protein H (CREBH) is a transcriptional regulator of hepatic autophagy in response to diet-induced NASH. The results showed that the upregulation of CREBH in lipid-overloaded hepatocytes improves cell damage, dysfunction of autophagic flux and associated abnormal accumulation of the autophagosome marker LC3-II and autophagic substrate p62. CREBH deficiency aggravated the dysfunctional autophagy and liver injury and even caused NASH-associated liver fibrosis. In addition, the changing trend of autolysosomes and lysosome membrane-associated protein (LAMP1) was consistent with the expression level of CREBH. This result indicated that CREBH might promote autophagic degradation by restoring the formation of autolysosomes, thereby improving the blocked autophagic flux. Moreover, we observed that CREBH inhibited the expression of Coronin 1a (Coro1a), an autophagosome-lysosome fusion-related gene, through transcriptional regulation. The overexpression of Coro1a in LO2 liver cells inhibited autophagic flux and elevated inflammatory cytokine levels upon palmitic acid (PA) stimulation. Overall, our findings provide new insights into the regulatory role of CREBH in the progression of NASH and reveal that Coro1a is a novel target gene of CREBH based on the autophagy pathway.
Collapse
Affiliation(s)
- Xiaoling Deng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Beibei Liu
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianqian Jiang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guixin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
9
|
Yuxiong W, Faping L, Bin L, Yanghe Z, Yao L, Yunkuo L, Yishu W, Honglan Z. Regulatory mechanisms of the cAMP-responsive element binding protein 3 (CREB3) family in cancers. Biomed Pharmacother 2023; 166:115335. [PMID: 37595431 DOI: 10.1016/j.biopha.2023.115335] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
The CREB3 family of proteins, encompassing CREB3 and its four homologs (CREB3L1, CREB3L2, CREB3L3, and CREB3L4), exerts pivotal control over cellular protein metabolism in response to unfolded protein reactions. Under conditions of endoplasmic reticulum stress, activation of the CREB3 family occurs through regulated intramembrane proteolysis within the endoplasmic reticulum membrane. Perturbations in the function and expression of the CREB3 family have been closely associated with the development of diverse diseases, with a particular emphasis on cancer. Recent investigations have shed light on the indispensable role played by CREB3 family members in modulating the onset and progression of various human cancers. This comprehensive review endeavors to provide an in-depth examination of the involvement of CREB3 family members in distinct human cancer types, accentuating their significance in the pathogenesis of cancer and the manifestation of malignant phenotypes.
Collapse
Affiliation(s)
- Wang Yuxiong
- Department of Urology II, The First Hospital of Jilin University, Changchun 130011, China
| | - Li Faping
- Department of Urology II, The First Hospital of Jilin University, Changchun 130011, China
| | - Liu Bin
- Department of Urology II, The First Hospital of Jilin University, Changchun 130011, China
| | - Zhang Yanghe
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130011, China
| | - Li Yao
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130011, China
| | - Li Yunkuo
- Department of Urology II, The First Hospital of Jilin University, Changchun 130011, China
| | - Wang Yishu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130011, China.
| | - Zhou Honglan
- Department of Urology II, The First Hospital of Jilin University, Changchun 130011, China,.
| |
Collapse
|
10
|
Zheng Y, Wang S, Wu J, Wang Y. Mitochondrial metabolic dysfunction and non-alcoholic fatty liver disease: new insights from pathogenic mechanisms to clinically targeted therapy. J Transl Med 2023; 21:510. [PMID: 37507803 PMCID: PMC10375703 DOI: 10.1186/s12967-023-04367-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is among the most widespread metabolic disease globally, and its associated complications including insulin resistance and diabetes have become threatening conditions for human health. Previous studies on non-alcoholic fatty liver disease (NAFLD) were focused on the liver's lipid metabolism. However, growing evidence suggests that mitochondrial metabolism is involved in the pathogenesis of NAFLD to varying degrees in several ways, for instance in cellular division, oxidative stress, autophagy, and mitochondrial quality control. Ultimately, liver function gradually declines as a result of mitochondrial dysfunction. The liver is unable to transfer the excess lipid droplets outside the liver. Therefore, how to regulate hepatic mitochondrial function to treat NAFLD has become the focus of current research. This review provides details about the intrinsic link of NAFLD with mitochondrial metabolism and the mechanisms by which mitochondrial dysfunctions contribute to NAFLD progression. Given the crucial role of mitochondrial metabolism in NAFLD progression, the application potential of multiple mitochondrial function improvement modalities (including physical exercise, diabetic medications, small molecule agonists targeting Sirt3, and mitochondria-specific antioxidants) in the treatment of NAFLD was evaluated hoping to provide new insights into NAFLD treatment.
Collapse
Affiliation(s)
- Youwei Zheng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Shiting Wang
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jialiang Wu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
11
|
Sun Y, Zhang Y, Zhang J, Chen YE, Jin JP, Zhang K, Mou H, Liang X, Xu J. XBP1-mediated transcriptional regulation of SLC5A1 in human epithelial cells in disease conditions. RESEARCH SQUARE 2023:rs.3.rs-3112506. [PMID: 37502997 PMCID: PMC10371076 DOI: 10.21203/rs.3.rs-3112506/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background sodium-dependent glucose cotransporter 1 and 2 (SGLT1/2) belong to the family of glucose transporters, encoded by SLC5A1 and SLC5A2, respectively. SGLT-2 is almost exclusively expressed in the renal proximal convoluted tubule cells. SGLT-1 is expressed in the kidneys but also in other organs throughout the body. Many SGLT inhibitor drugs have been developed based on the mechanism of blocking glucose (re)absorption mediated by SGLT1/2, and several have gained major regulatory agencies' approval for treating diabetes. Intriguingly these drugs are also effective in treating diseases beyond diabetes, for example heart failure and chronic kidney disease. We recently discovered that SGLT-1 is upregulated in the airway epithelial cells derived from patients of cystic fibrosis (CF), a devastating genetic disease affecting greater than 70,000 worldwide. Results in the present work, we show that the SGLT-1 upregulation is coupled with elevated endoplasmic reticulum (ER) stress response, indicated by activation of the primary ER stress senor inositol-requiring protein 1a (IRE1a) and the ER stress-induced transcription factor X-box binding protein 1 (XBP1), in CF epithelial cells, and in epithelial cells of other stress conditions. Through biochemistry experiments, we demonstrated that XBP1 acts as a transcription factor for SLC5A1 by directly binding to its promoter region. Targeting this ER stress → SLC5A1 axis by either the ER stress inhibitor Rapamycin or the SGLT-1 inhibitor Sotagliflozin was effective in attenuating the ER stress response and reducing the SGLT-1 levels in these cellular model systems. Conclusions the present work establishes a causal relationship between ER stress and SGLT-1 upregulation and provides a mechanistic explanation why SGLT inhibitor drugs benefit diseases beyond diabetes.
Collapse
Affiliation(s)
- Yifei Sun
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yihan Zhang
- The Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Jackson 1402, Boston, MA 02114, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Y. Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jian-Ping Jin
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Hongmei Mou
- The Mucosal Immunology & Biology Research Center, Massachusetts General Hospital, 55 Fruit Street, Jackson 1402, Boston, MA 02114, USA
| | - Xiubin Liang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
12
|
Mo P, Chen H, Jiang X, Hu F, Zhang F, Shan G, Chen W, Li S, Xu G. Effect of hepatic NPC1L1 on cholesterol gallstone disease and its mechanism. Heliyon 2023; 9:e15757. [PMID: 37159680 PMCID: PMC10163659 DOI: 10.1016/j.heliyon.2023.e15757] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/11/2023] Open
Abstract
Cholesterol gallstone disease (CGD) is associated with bile cholesterol supersaturation. The Niemann-Pick C1-like 1 (NPC1L1), the inhibitory target of ezetimibe (EZE), is a critical sterol transporter of cholesterol absorption. Intestinal NPC1L1 facilitates the absorption of cholesterol, whereas hepatic NPC1L1 promotes cholesterol uptake by hepatocytes and reduces bile cholesterol supersaturation. The potential of hepatic NPC1L1 to prevent CGD has yet to be established due to its absence in the mice model. In this study, we generated mice expressing hepatic NPC1L1 using adeno-associated virus (AAV) gene delivery. The biliary cholesterol saturations and gallstone formations were explored under chow diet and lithogenic diet (LD) with or without EZE treatment. The long-term (8-week) LD-fed AAV-mNPC1L1 mice exhibited no significant differences in biliary cholesterol saturation and gallstone formation compared to WT mice. EZE effectively prevented CGD in both WT and AAV-mNPC1L1 mice. Mechanistically, prolonged LD feeding induced the degradation of hepatic NPC1L1, whereas short-term (2-week) LD feeding preserved the expression of hepatic NPC1L1. In conclusion, our findings suggest that hepatic NPC1L1 is unable to prevent CGD, whereas EZE functions as an efficient bile cholesterol desaturator during CGD development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guoqiang Xu
- Corresponding author. Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou 310006, Zhejiang, China.
| |
Collapse
|
13
|
Cheng Y, Kang XZ, Chan P, Cheung PHH, Cheng T, Ye ZW, Chan CP, Yu CH, Jin DY. FACI is a novel clathrin adaptor protein 2-binding protein that facilitates low-density lipoprotein endocytosis. Cell Biosci 2023; 13:74. [PMID: 37072871 PMCID: PMC10114425 DOI: 10.1186/s13578-023-01023-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/27/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Cholesterol plays a vital role in multiple physiological processes. Cellular uptake of cholesterol is mediated primarily through endocytosis of low-density lipoprotein (LDL) receptor. New modifiers of this process remain to be characterized. Particularly, the role of fasting- and CREB-H-induced (FACI) protein in cholesterol homeostasis merits further investigation. METHODS Interactome profiling by proximity labeling and affinity purification - mass spectrometry was performed. Total internal reflection fluorescence microscopy and confocal immunofluorescence microscopy were used to analyze protein co-localization and interaction. Mutational analysis was carried out to define the domain and residues required for FACI localization and function. Endocytosis was traced by fluorescent cargos. LDL uptake in cultured cells and diet-induced hypercholesterolemia in mice were assessed. RESULTS FACI interacted with proteins critically involved in clathrin-mediated endocytosis, vesicle trafficking, and membrane cytoskeleton. FACI localized to clathrin-coated pits (CCP) on plasma membranes. FACI contains a conserved DxxxLI motif, which mediates its binding with the adaptor protein 2 (AP2) complex. Disruption of this motif of FACI abolished its CCP localization but didn't affect its association with plasma membrane. Cholesterol was found to facilitate FACI transport from plasma membrane to endocytic recycling compartment in a clathrin- and cytoskeleton-dependent manner. LDL endocytosis was enhanced in FACI-overexpressed AML12 cells but impaired in FACI-depleted HeLa cells. In vivo study indicated that hepatic FACI overexpression alleviated diet-induced hypercholesterolemia in mice. CONCLUSIONS FACI facilitates LDL endocytosis through its interaction with the AP2 complex.
Collapse
Affiliation(s)
- Yun Cheng
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
| | - Xiao-Zhuo Kang
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Pearl Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Pak-Hin Hinson Cheung
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Tao Cheng
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Zi-Wei Ye
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Cheng-Han Yu
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong.
| |
Collapse
|
14
|
Kim H, Song Z, Zhang R, Davies BSJ, Zhang K. A hepatokine derived from the ER protein CREBH promotes triglyceride metabolism by stimulating lipoprotein lipase activity. Sci Signal 2023; 16:eadd6702. [PMID: 36649378 PMCID: PMC10080946 DOI: 10.1126/scisignal.add6702] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
The endoplasmic reticulum (ER)-tethered, liver-enriched stress sensor CREBH is processed in response to increased energy demands or hepatic stress to release an amino-terminal fragment that functions as a transcription factor for hepatic genes encoding lipid and glucose metabolic factors. Here, we discovered that the carboxyl-terminal fragment of CREBH (CREBH-C) derived from membrane-bound, full-length CREBH was secreted as a hepatokine in response to fasting or hepatic stress. Phosphorylation of CREBH-C mediated by the kinase CaMKII was required for efficient secretion of CREBH-C through exocytosis. Lipoprotein lipase (LPL) mediates the lipolysis of circulating triglycerides for tissue uptake and is inhibited by a complex consisting of angiopoietin-like (ANGPTL) 3 and ANGPTL8. Secreted CREBH-C blocked the formation of ANGPTL3-ANGPTL8 complexes, leading to increased LPL activity in plasma and metabolic tissues in mice. CREBH-C administration promoted plasma triglyceride clearance and partitioning into peripheral tissues and mitigated hypertriglyceridemia and hepatic steatosis in mice fed a high-fat diet. Individuals with obesity had higher circulating amounts of CREBH-C than control individuals, and human CREBH loss-of-function variants were associated with dysregulated plasma triglycerides. These results identify a stress-induced, secreted protein fragment derived from CREBH that functions as a hepatokine to stimulate LPL activity and triglyceride homeostasis.
Collapse
Affiliation(s)
- Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Brandon S. J. Davies
- Department of Biochemistry, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
15
|
Barrabi C, Zhang K, Liu M, Chen X. Pancreatic beta cell ER export in health and diabetes. Front Endocrinol (Lausanne) 2023; 14:1155779. [PMID: 37152949 PMCID: PMC10160654 DOI: 10.3389/fendo.2023.1155779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
In the secretory pathway of the pancreatic beta cell, proinsulin and other secretory granule proteins are first produced in the endoplasmic reticulum (ER). Beta cell ER homeostasis is vital for normal beta cell functions and is maintained by the delicate balance between protein synthesis, folding, export and degradation. Disruption of ER homeostasis leads to beta cell death and diabetes. Among the four components to maintain ER homeostasis, the role of ER export in insulin biogenesis or beta cell survival was not well-understood. COPII (coat protein complex II) dependent transport is a conserved mechanism for most cargo proteins to exit ER and transport to Golgi apparatus. Emerging evidence began to reveal a critical role of COPII-dependent ER export in beta cells. In this review, we will first discuss the basic components of the COPII transport machinery, the regulation of cargo entry and COPII coat assembly in mammalian cells, and the general concept of receptor-mediated cargo sorting in COPII vesicles. On the basis of these general discussions, the current knowledge and recent developments specific to the beta cell COPII dependent ER export are summarized under normal and diabetic conditions.
Collapse
Affiliation(s)
- Cesar Barrabi
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, United States
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuequn Chen
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, United States
- *Correspondence: Xuequn Chen,
| |
Collapse
|
16
|
Wu Q, Liang X, Hou X, Song Z, Bouhamdan M, Qiu Y, Koike Y, Rajagopalan C, Wei HG, Jiang H, Hish G, Zhang J, Chen YE, Jin JP, Xu J, Zhang K, Sun F. Cystic fibrosis rabbits develop spontaneous hepatobiliary lesions and CF-associated liver disease (CFLD)-like phenotypes. PNAS NEXUS 2023; 2:pgac306. [PMID: 36712930 PMCID: PMC9832953 DOI: 10.1093/pnasnexus/pgac306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disease affecting multiple organs. Approximately 30% CF patients develop CF-related liver disease (CFLD), which is the third most common cause of morbidity and mortality of CF. CFLD is progressive, and many of the severe forms eventually need liver transplantation. The mechanistic studies and therapeutic interventions to CFLD are unfortunately very limited. Utilizing the CRISPR/Cas9 technology, we recently generated CF rabbits by introducing mutations to the rabbit CF transmembrane conductance regulator (CFTR) gene. Here we report the liver phenotypes and mechanistic insights into the liver pathogenesis in these animals. CF rabbits develop spontaneous hepatobiliary lesions and abnormal biliary secretion accompanied with altered bile acid profiles. They exhibit nonalcoholic steatohepatitis (NASH)-like phenotypes, characterized by hepatic inflammation, steatosis, and fibrosis, as well as altered lipid profiles and diminished glycogen storage. Mechanistically, our data reveal that multiple stress-induced metabolic regulators involved in hepatic lipid homeostasis were up-regulated in the livers of CF-rabbits, and that endoplasmic reticulum (ER) stress response mediated through IRE1α-XBP1 axis as well as NF-κB- and JNK-mediated inflammatory responses prevail in CF rabbit livers. These findings show that CF rabbits manifest many CFLD-like phenotypes and suggest targeting hepatic ER stress and inflammatory pathways for potential CFLD treatment.
Collapse
Affiliation(s)
- Qingtian Wu
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xiubin Liang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Xia Hou
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Mohamad Bouhamdan
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yining Qiu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yui Koike
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Carthic Rajagopalan
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hong-Guang Wei
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Hong Jiang
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Gerry Hish
- Laboratory Animal Resources, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Fei Sun
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
17
|
Mo P, Chen H, Jiang X, Hu F, Zhang F, Shan G, Chen W, Li S, Li Y, Xu G. FGF15 promotes hepatic NPC1L1 degradation in lithogenic diet-fed mice. Lipids Health Dis 2022; 21:97. [PMID: 36209166 PMCID: PMC9547418 DOI: 10.1186/s12944-022-01709-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cholesterol gallstone disease (CGD) is accompanied by biliary cholesterol supersaturation. Hepatic Niemann-Pick C1-like 1 (NPC1L1), which is present in humans but not in wild-type (WT) mice, promotes hepatocyte cholesterol uptake and decreases biliary cholesterol supersaturation. In contrast, intestinal NPC1L1 promotes intestinal cholesterol absorption, increasing biliary cholesterol supersaturation. Ezetimibe (EZE) can inhibit both hepatic and intestinal NPC1L1. However, whether hepatic NPC1L1 can affect CGD progress remains unknown. METHODS Mice expressing hepatic NPC1L1 (NPC1L1hepatic-OE mice) were generated using Adeno-associated viruses (AAV) gene delivery. The protein level and function of hepatic NPC1L1 were examined under chow diet, high fat-cholesterol diet (HFCD), and lithogenic diet (LD) feeding. Gallstone formation rates were examined with or without EZE treatment. Fibroblast growth factor 15 (FGF15) treatment and inhibition of fibroblast growth factor receptor 4 (FGFR4) were applied to verify the mechanism of hepatic NPC1L1 degradation. RESULTS The HFCD-fed NPC1L1hepatic-OE mice retained the biliary cholesterol desaturation function of hepatic NPC1L1, whereas EZE treatment decreased biliary cholesterol saturation and did not cause CGD. The ubiquitination and degradation of hepatic NPC1L1 were discovered in LD-fed NPC1L1hepatic-OE mice. Treatment of FGF15 during HFCD feeding and inhibition of FGFR4 during LD feeding could affect the protein level and function of hepatic NPC1L1. CONCLUSIONS LD induces the ubiquitination and degradation of hepatic NPC1L1 via the FGF15-FGFR4 pathway. EZE may act as an effective preventative agent for CGD.
Collapse
Affiliation(s)
- Pingfan Mo
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Hongtan Chen
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Xin Jiang
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Fengling Hu
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Fenming Zhang
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Guodong Shan
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Wenguo Chen
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Sha Li
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China
| | - Yiqiao Li
- Urology& Nephrology Center, Department of Nephrology, Zhejiang Provincial People's Hospital and Hangzhou Medical College Affiliated People's Hospital, 158 Shangtang Road, Hangzhou, 310014, Zhejiang, China.
| | - Guoqiang Xu
- Department of Gastroenterology, Zhejiang University School of Medicine, The First Affiliated Hospital, 79 Qingchun Road, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
18
|
Kim H, Fu Z, Yang Z, Song Z, Shamsa EH, Yumnamcha T, Sun S, Liu W, Ibrahim AS, Qi NR, Zhang R, Zhang K. The mitochondrial NAD kinase functions as a major metabolic regulator upon increased energy demand. Mol Metab 2022; 64:101562. [PMID: 35944895 PMCID: PMC9403569 DOI: 10.1016/j.molmet.2022.101562] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE The mitochondrial nicotinamide adenine dinucleotide (NAD) kinase (MNADK) mediates de novo mitochondrial NADP biosynthesis by catalyzing the phosphorylation of NAD to yield NADP. In this study, we investigated the function and mechanistic basis by which MNADK regulates metabolic homeostasis. METHODS Generalized gene set analysis by aggregating human patient genomic databases, metabolic studies with genetically engineered animal models, mitochondrial bioenergetic analysis, as well as gain- and loss- of-function studies were performed to address the functions and mechanistic basis by which MNADK regulates energy metabolism and redox state associated with metabolic disease. RESULTS Human MNADK common gene variants or decreased expression of the gene are significantly associated with the occurrence of type-2 diabetes, non-alcoholic fatty liver disease (NAFLD), or hepatocellular carcinoma (HCC). Ablation of the MNADK gene in mice led to decreased fat oxidation, coincident with increased respiratory exchange ratio (RER) and decreased energy expenditure upon energy demand triggered by endurance exercise or fasting. On an atherogenic high-fat diet (HFD), MNADK-null mice exhibited hepatic insulin resistance and glucose intolerance, indicating a type-2 diabetes-like phenotype in the absence of MNADK. MNADK deficiency led to a decrease in mitochondrial NADP(H) but an increase in cellular reactive oxygen species (ROS) in mouse livers. Consistently, protein levels of the major metabolic regulators or enzymes were decreased, while their acetylation modifications were increased in the livers of MNADK-null mice. Feeding mice with a HFD caused S-nitrosylation (SNO) modification, a posttranslational modification that represses protein activities, on MNADK protein in the liver. Reconstitution of an SNO-resistant MNADK variant, MNADK-S193, into MNADK-null mice mitigated hepatic steatosis induced by HFD. CONCLUSION MNADK, the only known mammalian mitochondrial NAD kinase, plays important roles in preserving energy homeostasis to mitigate the risk of metabolic disorders.
Collapse
Affiliation(s)
- Hyunbae Kim
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA
| | - Zhiyao Fu
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA
| | - Zhao Yang
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA
| | | | - Thangal Yumnamcha
- Departments of Ophthalmology Visual Anatomical Science and pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Shengyi Sun
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA
| | - Wanqing Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Ahmed S Ibrahim
- Departments of Ophthalmology Visual Anatomical Science and pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Nathan R Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA; Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Detroit, MI 48201, USA; Department of Biochemistry, Microbiology, and Immunology, Detroit, MI 48201, USA.
| |
Collapse
|
19
|
Zhang J, Zhao Y, Wang S, Li G, Xu K. CREBH alleviates mitochondrial oxidative stress through SIRT3 mediating deacetylation of MnSOD and suppression of Nlrp3 inflammasome in NASH. Free Radic Biol Med 2022; 190:28-41. [PMID: 35926687 DOI: 10.1016/j.freeradbiomed.2022.07.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/23/2022] [Accepted: 07/24/2022] [Indexed: 02/07/2023]
Abstract
Lipotoxicity and unresolved oxidative stress are key drivers of metabolic inflammation in nonalcoholic steatohepatitis (NASH). cAMP-response element binding protein H(CREBH) is a liver-specific transcription factor and regulates the glucose and lipid metabolism of NASH. However, its role in mitochondrial oxidative stress and its association with sirtuin 3 (SIRT3), a master regulator of deacetylation for mitochondrial proteins, remains elusive. In this study, AML-12 cells were treated with palmitic acid to imitate the pathological changes of NASH in vitro and 8-week-old male C57BL/6J mice were fed with a high-fat (HF) diet or a methionine-choline-deficient (MCD) diet to build the widely accepted in vivo model of NASH. We found that lipid overload induced mitochondrial oxidative stress and stimulated the expression of CREBH and SIRT3. CREBH overexpression alleviated the mitochondrial oxidative stress. Moreover, CREBH promoted SIRT3 expression, which regulated the deacetylation of manganese superoxide dismutase (MnSOD) and inhibited NOD-Like Receptor Pyrin Domain Containing 3 (Nlrp3) inflammasome activation whereas suppression of SIRT3 damaged the protecting ability of CREBH in mitochondrial oxidative stress. CREBH knockout mice were highly susceptible to HF and MCD diet-induced NASH with more severe oxidative stress. Collectively, our results firstly provided the support that CREBH could serve as a protective factor in the progression of NASH by regulating the acetylation of MnSOD and the activation of Nlrp3 inflammasome through SIRT3. These results suggest that CREBH might be a valuable therapeutic candidate for NASH.
Collapse
Affiliation(s)
- Junli Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yajuan Zhao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuhan Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guixin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
20
|
Ramanathan R, Ali AH, Ibdah JA. Mitochondrial Dysfunction Plays Central Role in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms23137280. [PMID: 35806284 PMCID: PMC9267060 DOI: 10.3390/ijms23137280] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 12/04/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global pandemic that affects one-quarter of the world’s population. NAFLD includes a spectrum of progressive liver disease from steatosis to nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis and can be complicated by hepatocellular carcinoma. It is strongly associated with metabolic syndromes, obesity, and type 2 diabetes, and it has been shown that metabolic dysregulation is central to its pathogenesis. Recently, it has been suggested that metabolic- (dysfunction) associated fatty liver disease (MAFLD) is a more appropriate term to describe the disease than NAFLD, which puts increased emphasis on the important role of metabolic dysfunction in its pathogenesis. There is strong evidence that mitochondrial dysfunction plays a significant role in the development and progression of NAFLD. Impaired mitochondrial fatty acid oxidation and, more recently, a reduction in mitochondrial quality, have been suggested to play a major role in NAFLD development and progression. In this review, we provide an overview of our current understanding of NAFLD and highlight how mitochondrial dysfunction contributes to its pathogenesis in both animal models and human subjects. Further we discuss evidence that the modification of mitochondrial function modulates NAFLD and that targeting mitochondria is a promising new avenue for drug development to treat NAFLD/NASH.
Collapse
Affiliation(s)
- Raghu Ramanathan
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA; (R.R.); (A.H.A.)
- Harry S. Truman Memorial Veterans Medical Center, Columbia, MO 65201, USA
| | - Ahmad Hassan Ali
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA; (R.R.); (A.H.A.)
- Harry S. Truman Memorial Veterans Medical Center, Columbia, MO 65201, USA
| | - Jamal A. Ibdah
- Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA; (R.R.); (A.H.A.)
- Harry S. Truman Memorial Veterans Medical Center, Columbia, MO 65201, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
- Correspondence: ; Tel.: +573-882-7349; Fax: +573-884-4595
| |
Collapse
|
21
|
Zhang K. Stress-induced Regulators of Intestinal Fat Absorption. Cell Mol Gastroenterol Hepatol 2022; 13:1469-1470. [PMID: 35189121 PMCID: PMC9043301 DOI: 10.1016/j.jcmgh.2022.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/10/2022]
Affiliation(s)
- Kezhong Zhang
- Center for Molecular Medicine and Genetics, Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan.
| |
Collapse
|
22
|
McCann MA, Li Y, Muñoz M, Gil V, Qiang G, Cordoba-Chacon J, Blüher M, Duncan S, Liew CW. Adipose expression of CREB3L3 modulates body weight during obesity. Sci Rep 2021; 11:19400. [PMID: 34588527 PMCID: PMC8481296 DOI: 10.1038/s41598-021-98627-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 08/30/2021] [Indexed: 11/08/2022] Open
Abstract
We found the hepatic transcription factor Cyclic-AMP Responsive Element Binding Protein 3-like-3 (CREB3L3) to be expressed in adipose tissue, and selectively downregulated in the more metabolically protective subcutaneous adipose tissue in obese mice and humans. We sought to elucidate the specific role of this factor in adipose biology. CREB3L3 fat-specific knockout mice were fed a high-fat diet to induce obesity and metabolic dysfunction. Additionally, we injected a flip-excision adeno-associated virus directly into the subcutaneous inguinal adipose tissue of Adiponectin-Cre mice to create a depot-specific overexpression model for further assessment. Fat-specific ablation of CREB3L3 enhanced weight gain and insulin resistance following high-fat feeding, as fat-specific knockout mice expended less energy and possessed more inflammatory adipose tissue. Conversely, inguinal fat CREB3L3 overexpression deterred diet-induced obesity and ameliorated metabolic dysfunction. Together, this study highlights the relevance of CREB3L3 in obese adipose tissue and demonstrates its role as a powerful body weight modulator.
Collapse
Affiliation(s)
- Maximilian A McCann
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Yanliang Li
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Marcos Muñoz
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Victoria Gil
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Guifen Qiang
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jose Cordoba-Chacon
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Stephen Duncan
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Chong Wee Liew
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Physiology & Biophysics, College of Medicine, University of Illinois at Chicago, 835 S Wolcott Ave, M/C 901, MSB, E-202, Chicago, IL, 60612, USA.
| |
Collapse
|
23
|
CREBH Systemically Regulates Lipid Metabolism by Modulating and Integrating Cellular Functions. Nutrients 2021; 13:nu13093204. [PMID: 34579081 PMCID: PMC8472586 DOI: 10.3390/nu13093204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Cyclic AMP-responsive element-binding protein H (CREBH, encoded by CREB3L3) is a membrane-bound transcriptional factor expressed in the liver and small intestine. The activity of CREBH is regulated not only at the transcriptional level but also at the posttranslational level. CREBH governs triglyceride metabolism in the liver by controlling gene expression, with effects including the oxidation of fatty acids, lipophagy, and the expression of apolipoproteins related to the lipoprotein lipase activation and suppression of lipogenesis. The activation and functions of CREBH are controlled in response to the circadian rhythm. On the other hand, intestinal CREBH downregulates the absorption of lipids from the diet. CREBH deficiency in mice leads to severe hypertriglyceridemia and fatty liver in the fasted state and while feeding a high-fat diet. Therefore, when crossing CREBH knockout (KO) mice with an atherosclerosis model, low-density lipoprotein receptor KO mice, these mice exhibit severe atherosclerosis. This phenotype is seen in both liver- and small intestine-specific CREBH KO mice, suggesting that CREBH controls lipid homeostasis in an enterohepatic interaction. This review highlights that CREBH has a crucial role in systemic lipid homeostasis to integrate cellular functions related to lipid metabolism.
Collapse
|
24
|
Yang Z, Roth K, Agarwal M, Liu W, Petriello MC. The transcription factors CREBH, PPARa, and FOXO1 as critical hepatic mediators of diet-induced metabolic dysregulation. J Nutr Biochem 2021; 95:108633. [PMID: 33789150 PMCID: PMC8355060 DOI: 10.1016/j.jnutbio.2021.108633] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/31/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
The liver is a critical mediator of lipid and/or glucose homeostasis and is a primary organ involved in dynamic changes during feeding and fasting. Additionally, hepatic-centric pathways are prone to dysregulation during pathophysiological states including metabolic syndrome (MetS) and non-alcoholic fatty liver disease. Omics platforms and GWAS have elucidated genes related to increased risk of developing MetS and related disorders, but mutations in these metabolism-related genes are rare and cannot fully explain the increasing prevalence of MetS-related pathologies worldwide. Complex interactions between diet, lifestyle, environmental factors, and genetic predisposition jointly determine inter-individual variability of disease risk. Given the complexity of these interactions, researchers have focused on master regulators of metabolic responses incorporating and mediating the impact of multiple environmental cues. Transcription factors are DNA binding, terminal executors of signaling pathways that modulate the cellular responses to complex metabolic stimuli and are related to the control of hepatic lipid and glucose homeostasis. Among numerous hepatic transcription factors involved in regulating metabolism, three emerge as key players in transducing nutrient sensing, which are dysregulated in MetS-related perturbations in both clinical and preclinical studies: cAMP Responsive Element Binding Protein 3 Like 3 (CREB3L3), Peroxisome Proliferator Activated Receptor Alpha (PPAR), and Forkhead Box O1 (FOXO1). Additionally, these three transcription factors appear to be amenable to dietary and/or nutrient-based therapies, being potential targets of nutritional therapy. In this review we aim to describe the activation, regulation, and impact of these transcription factors in the context of metabolic homeostasis. We also summarize their perspectives in MetS and nutritional therapies.
Collapse
Affiliation(s)
- Zhao Yang
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA
| | - Katherine Roth
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA
| | - Manisha Agarwal
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Wanqing Liu
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Wayne State University, Detroit, MI, USA
| | - Michael C Petriello
- Institute of Environmental Health Sciences (IEHS), Wayne State University, Detroit, MI, USA; Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
25
|
Kang M, Han SK, Kim S, Park S, Jo Y, Kang H, Ko J. Role of small leucine zipper protein in hepatic gluconeogenesis and metabolic disorder. J Mol Cell Biol 2021; 13:361-373. [PMID: 33355643 PMCID: PMC8373270 DOI: 10.1093/jmcb/mjaa069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/03/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Hepatic gluconeogenesis is the central pathway for glucose generation in the body. The imbalance between glucose synthesis and uptake leads to metabolic diseases such as obesity, diabetes, and cardiovascular diseases. Small leucine zipper protein (sLZIP) is an isoform of LZIP and it mainly functions as a transcription factor. Although sLZIP is known to regulate the transcription of genes involved in various cellular processes, the role of sLZIP in hepatic glucose metabolism is not known. In this study, we investigated the regulatory role of sLZIP in hepatic gluconeogenesis and its involvement in metabolic disorder. We found that sLZIP expression was elevated during glucose starvation, leading to the promotion of phosphoenolpyruvate carboxylase and glucose-6-phosphatase expression in hepatocytes. However, sLZIP knockdown suppressed the expression of the gluconeogenic enzymes under low glucose conditions. sLZIP also enhanced glucose production in the human liver cells and mouse primary hepatic cells. Fasting-induced cyclic adenosine monophosphate impeded sLZIP degradation. Results of glucose and pyruvate tolerance tests showed that sLZIP transgenic mice exhibited abnormal blood glucose metabolism. These findings suggest that sLZIP is a novel regulator of gluconeogenic enzyme expression and plays a role in blood glucose homeostasis during starvation.
Collapse
Affiliation(s)
- Minsoo Kang
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Sun Kyoung Han
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Suhyun Kim
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Sungyeon Park
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Yerin Jo
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Hyeryung Kang
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Jesang Ko
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| |
Collapse
|
26
|
Nakagawa Y, Kumagai K, Han SI, Mizunoe Y, Araki M, Mizuno S, Ohno H, Matsuo K, Yamada Y, Kim JD, Miyamoto T, Sekiya M, Konishi M, Itoh N, Matsuzaka T, Takahashi S, Sone H, Shimano H. Starvation-induced transcription factor CREBH negatively governs body growth by controlling GH signaling. FASEB J 2021; 35:e21663. [PMID: 34042217 DOI: 10.1096/fj.202002784rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/13/2022]
Abstract
cAMP responsive element-binding protein H (CREBH) is a hepatic transcription factor to be activated during fasting. We generated CREBH knock-in flox mice, and then generated liver-specific CREBH transgenic (CREBH L-Tg) mice in an active form. CREBH L-Tg mice showed a delay in growth in the postnatal stage. Plasma growth hormone (GH) levels were significantly increased in CREBH L-Tg mice, but plasma insulin-like growth factor 1 (IGF1) levels were significantly decreased, indicating GH resistance. In addition, CREBH overexpression significantly increased hepatic mRNA and plasma levels of FGF21, which is thought to be as one of the causes of growth delay. However, the additional ablation of FGF21 in CREBH L-Tg mice could not correct GH resistance at all. CREBH L-Tg mice sustained GH receptor (GHR) reduction and the increase of IGF binding protein 1 (IGFBP1) in the liver regardless of FGF21. As GHR is a first step in GH signaling, the reduction of GHR leads to impairment of GH signaling. These data suggest that CREBH negatively regulates growth in the postnatal growth stage via various pathways as an abundant energy response by antagonizing GH signaling.
Collapse
Affiliation(s)
- Yoshimi Nakagawa
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan.,Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Kae Kumagai
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Song-Iee Han
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yuhei Mizunoe
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Masaya Araki
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroshi Ohno
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuya Matsuo
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yasunari Yamada
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Jun-Dal Kim
- Division of Complex Biosystem Research, Department of Research and Development, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Takafumi Miyamoto
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Morichika Konishi
- Department of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Science, Kyoto University, Kyoto, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Transborder Medical Research Center (TMRC), University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Transborder Medical Research Center (TMRC), University of Tsukuba, Tsukuba, Japan.,Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hirohito Sone
- Faculty of Medicine, Department of Hematology, Endocrinology and Metabolism, Niigata University, Niigata, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.,Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
27
|
Kim H, Wei J, Song Z, Mottillo E, Samavati L, Zhang R, Li L, Chen X, Jena BP, Lin JD, Fang D, Zhang K. Regulation of hepatic circadian metabolism by the E3 ubiquitin ligase HRD1-controlled CREBH/PPARα transcriptional program. Mol Metab 2021; 49:101192. [PMID: 33592335 PMCID: PMC7966871 DOI: 10.1016/j.molmet.2021.101192] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE The endoplasmic reticulum (ER)-resident E3 ligase HRD1 and its co-activator Sel1L are major components of ER-associated degradation (ERAD) machinery. Here, we investigated the molecular mechanism and functional significance underlying the circadian regulation of HRD1/Sel1L-mediated protein degradation program in hepatic energy metabolism. METHODS Genetically engineered animal models as well as gain- and loss-of-function studies were employed to address the circadian regulatory mechanism and functional significance. Gene expression, transcriptional activation, protein-protein interaction, and animal metabolic phenotyping analyses were performed to dissect the molecular network and metabolic pathways. RESULTS Hepatic HRD1 and Sel1L expression exhibits circadian rhythmicity that is controlled by the ER-tethered transcriptional activator CREBH, the nuclear receptor peroxisome proliferator-activated receptor α (PPARα), and the core clock oscillator BMAL1 in mouse livers. HRD1/Sel1L mediates polyubiquitination and degradation of the CREBH protein across the circadian cycle to modulate rhythmic expression of the genes encoding the rate-limiting enzymes or regulators in fatty acid (FA) oxidation, triglyceride (TG) lipolysis, lipophagy, and gluconeogenesis. HRD1 liver-specific knockout (LKO) mice displayed increased expression of the genes involved in lipid and glucose metabolism and impaired circadian profiles of circulating TG, FA, and glucose due to overproduction of CREBH. The circadian metabolic activities of HRD1 LKO mice were inversely correlated with those of CREBH KO mice. Suppressing CREBH overproduction in the livers of HRD1 LKO mice restored the diurnal levels of circulating TG and FA of HRD1 LKO mice. CONCLUSION Our work revealed a key circadian-regulated molecular network through which the E3 ubiquitin ligase HRD1 and its co-activator Sel1L regulate hepatic circadian metabolism.
Collapse
Affiliation(s)
- Hyunbae Kim
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Emilio Mottillo
- Hypertension and Vascular Research Division, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Lobelia Samavati
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Li Li
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xuequn Chen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Bhanu P Jena
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA; NanoBioScience Institute, Wayne State University, Detroit, MI 48201, USA
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA; Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA; NanoBioScience Institute, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
28
|
Krumm CS, Xu X, Bare CJ, Holman CD, Kersten S, Dow LE, Lee AH, Cohen DE. Inducible hepatic expression of CREBH mitigates diet-induced obesity, insulin resistance, and hepatic steatosis in mice. J Biol Chem 2021; 297:100815. [PMID: 34023388 PMCID: PMC8246594 DOI: 10.1016/j.jbc.2021.100815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Cyclic AMP-responsive element-binding protein H (CREBH encoded by Creb3l3) is a transcription factor that regulates the expression of genes that control lipid and glucose metabolism as well as inflammation. CREBH is upregulated in the liver under conditions of overnutrition, and mice globally lacking the gene (CREBH-/-) are highly susceptible to diet-induced obesity, insulin resistance, and hepatic steatosis. The net protective effects of CREBH have been attributed in large part to the activities of fibroblast growth factor (Fgf)-21 (Fgf21), a target gene that promotes weight loss, improves glucose homeostasis, and reduces hepatic lipid accumulation. To explore the possibility that activation of the CREBH-Fgf21 axis could ameliorate established effects of high-fat feeding, we generated an inducible transgenic hepatocyte-specific CREBH overexpression mouse model (Tg-rtTA). Acute overexpression of CREBH in livers of Tg-rtTA mice effectively reversed diet-induced obesity, insulin resistance, and hepatic steatosis. These changes were associated with increased activities of thermogenic brown and beige adipose tissues in Tg-rtTA mice, leading to reductions in fat mass, along with enhanced insulin sensitivity and glucose tolerance. Genetically silencing Fgf21 in Tg-rtTA mice abrogated the CREBH-mediated reductions in body weight loss, but only partially reversed the observed improvements in glucose metabolism. These findings reveal that the protective effects of CREBH activation may be leveraged to mitigate diet-induced obesity and associated metabolic abnormalities in both Fgf21-dependent and Fgf21-independent pathways.
Collapse
Affiliation(s)
- Christopher S Krumm
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Xu Xu
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Curtis J Bare
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Corey D Holman
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Lukas E Dow
- Division of Hematology & Medical Oncology, Joan & Sanford I. Weill Department of Medicine, Sandra and Edward Meyer Cancer Center, Weill Cornell Medical College, New York, New York, USA
| | - Ann-Hwee Lee
- Department of Pathology & Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - David E Cohen
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, New York, USA.
| |
Collapse
|
29
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) was defined in 1980 and has the same histological characteristics as alcoholic liver disease except for alcohol consumption. After 40 years, the understanding of this disease is still imperfect. Without specific drugs available for treatment, the number of patients with NAFLD is increasing rapidly, and NAFLD currently affects more than one-quarter of the global population. NAFLD is mostly caused by a sedentary lifestyle and excessive energy intake of fat and sugar. To ameliorate or avoid NAFLD, people commonly replace high-fat foods with high-carbohydrate foods (especially starchy carbohydrates) as a way to reduce caloric intake and reach satiety. However, there are few studies that concentrate on the effect of carbohydrate intake on liver metabolism in patients with NAFLD, much fewer than the studies on fat intake. Besides, most of these studies are not systematic, which has made identification of the mechanism difficult. In this review, we collected and analysed data from studies on human and animal models and, surprisingly, found that carbohydrates and liver steatosis could be linked by inflammation. This review not only describes the effects of carbohydrates on NAFLD and body lipid metabolism but also analyses and predicts possible molecular pathways of carbohydrates in liver lipid synthesis that involve inflammation. Furthermore, the limitations of recent research and possible targets for regulating inflammation and lipogenesis are discussed. This review describes the effects of starchy carbohydrates, a nutrient signal, on NAFLD from the perspective of inflammation.
Collapse
|
30
|
Cui A, Ding D, Li Y. Regulation of Hepatic Metabolism and Cell Growth by the ATF/CREB Family of Transcription Factors. Diabetes 2021; 70:653-664. [PMID: 33608424 PMCID: PMC7897342 DOI: 10.2337/dbi20-0006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022]
Abstract
The liver is a major metabolic organ that regulates the whole-body metabolic homeostasis and controls hepatocyte proliferation and growth. The ATF/CREB family of transcription factors integrates nutritional and growth signals to the regulation of metabolism and cell growth in the liver, and deregulated ATF/CREB family signaling is implicated in the progression of type 2 diabetes, nonalcoholic fatty liver disease, and cancer. This article focuses on the roles of the ATF/CREB family in the regulation of glucose and lipid metabolism and cell growth and its importance in liver physiology. We also highlight how the disrupted ATF/CREB network contributes to human diseases and discuss the perspectives of therapeutically targeting ATF/CREB members in the clinic.
Collapse
Affiliation(s)
- Aoyuan Cui
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
31
|
Wei J, Fang D. Endoplasmic Reticulum Stress Signaling and the Pathogenesis of Hepatocarcinoma. Int J Mol Sci 2021; 22:ijms22041799. [PMID: 33670323 PMCID: PMC7918477 DOI: 10.3390/ijms22041799] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC), also known as hepatoma, is a primary malignancy of the liver and the third leading cause of cancer mortality globally. Although much attention has focused on HCC, its pathogenesis remains largely obscure. The endoplasmic reticulum (ER) is a cellular organelle important for regulating protein synthesis, folding, modification and trafficking, and lipid metabolism. ER stress occurs when ER homeostasis is disturbed by numerous environmental, physiological, and pathological challenges. In response to ER stress due to misfolded/unfolded protein accumulation, unfolded protein response (UPR) is activated to maintain ER function for cell survival or, in cases of excessively severe ER stress, initiation of apoptosis. The liver is especially susceptible to ER stress given its protein synthesis and detoxification functions. Experimental data suggest that ER stress and unfolded protein response are involved in HCC development, aggressiveness and response to treatment. Herein, we highlight recent findings and provide an overview of the evidence linking ER stress to the pathogenesis of HCC.
Collapse
|
32
|
Aras S, Purandare N, Gladyck S, Somayajulu-Nitu M, Zhang K, Wallace DC, Grossman LI. Mitochondrial Nuclear Retrograde Regulator 1 (MNRR1) rescues the cellular phenotype of MELAS by inducing homeostatic mechanisms. Proc Natl Acad Sci U S A 2020; 117:32056-32065. [PMID: 33257573 PMCID: PMC7749287 DOI: 10.1073/pnas.2005877117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MNRR1 (CHCHD2) is a bi-organellar regulator of mitochondrial function that directly activates cytochrome c oxidase in the mitochondria and functions in the nucleus as a transcriptional activator for hundreds of genes. Since MNRR1 depletion contains features of a mitochondrial disease phenotype, we evaluated the effects of forced expression of MNRR1 on the mitochondrial disease MELAS (mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes) syndrome. MELAS is a multisystem encephalomyopathy disorder that can result from a heteroplasmic mutation in the mitochondrial DNA (mtDNA; m.3243A > G) at heteroplasmy levels of ∼50 to 90%. Since cybrid cell lines with 73% m.3243A > G heteroplasmy (DW7) display a significant reduction in MNRR1 levels compared to the wild type (0% heteroplasmy) (CL9), we evaluated the effects of MNRR1 levels on mitochondrial functioning. Overexpression of MNRR1 in DW7 cells induces the mitochondrial unfolded protein response (UPRmt), autophagy, and mitochondrial biogenesis, thereby rescuing the mitochondrial phenotype. It does so primarily as a transcription activator, revealing this function to be a potential therapeutic target. The role of MNRR1 in stimulating UPRmt, which is blunted in MELAS cells, was surprising and further investigation uncovered that under conditions of stress the import of MNRR1 into the mitochondria was blocked, allowing the protein to accumulate in the nucleus to enhance its transcription function. In the mammalian system, ATF5, has been identified as a mediator of UPRmt MNRR1 knockout cells display an ∼40% reduction in the protein levels of ATF5, suggesting that MNRR1 plays an important role upstream of this known mediator of UPRmt.
Collapse
Affiliation(s)
- Siddhesh Aras
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Neeraja Purandare
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Stephanie Gladyck
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Mallika Somayajulu-Nitu
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104;
- Department of Pediatrics, Division of Human Genetics, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lawrence I Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201;
| |
Collapse
|
33
|
Pyun DH, Kim TJ, Kim MJ, Hong SA, Abd El-Aty AM, Jeong JH, Jung TW. Endogenous metabolite, kynurenic acid, attenuates nonalcoholic fatty liver disease via AMPK/autophagy- and AMPK/ORP150-mediated signaling. J Cell Physiol 2020; 236:4902-4912. [PMID: 33283879 DOI: 10.1002/jcp.30199] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/03/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Endoplasmic reticulum (ER) stress plays a causative role in the development of nonalcoholic fatty liver disease (NAFLD). Kynurenic acid (KA) is a tryptophan metabolite that has been shown to exert anti-inflammatory effects in macrophages and endothelial cells. However, the role of KA in ER stress-associated development of NAFLD has not been fully explored. In the current study, we observed decreased KA levels in the serum of obese subjects. Treated hepatocytes with KA attenuated palmitate-induced lipid accumulation and downregulated lipogenesis-associated genes as well as ER stress markers in a dose-dependent manner. Furthermore, KA augmented AMP-activated protein kinase (AMPK) phosphorylation, oxygen-regulated protein 150 (ORP150) expression, and autophagy markers. The small interfering RNA-mediated suppression of AMPK and ORP150, or 3-methyladenine also abrogated the effects of KA on ER stress and lipid accumulation in hepatocytes. In accordance with in vitro observations, KA administration to mice fed a high-fat diet ameliorated hepatic lipid accumulation and decreased the expression of lipogenic genes as well as ER stress. Moreover, KA treatment increased hepatic AMPK phosphorylation, ORP150 expression, and autophagy related markers in mouse livers. Knockdown of AMPK using in vivo transfection mitigated the effects of KA on hepatic steatosis and ER stress as well as autophagy and ORP150 expression. These results suggest that KA ameliorates hepatic steatosis via the AMPK/autophagy- and AMPK/ORP150-mediated suppression of ER stress. In sum, KA might be used as a promising therapeutic agent for treatment of NAFLD.
Collapse
Affiliation(s)
- Do Hyeon Pyun
- Department of Pharmacology, Chung-Ang University, Seoul, Republic of Korea
| | - Tae Jin Kim
- Department of Pharmacology, Chung-Ang University, Seoul, Republic of Korea
| | - Myeong Jun Kim
- Department of Pharmacology, Chung-Ang University, Seoul, Republic of Korea
| | - Soon Auck Hong
- Department of Pathology, Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.,State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Shandong Academy of Science, Qilu University of Technology, Jinan, China.,Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Ji Hoon Jeong
- Department of Pharmacology, Chung-Ang University, Seoul, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
34
|
Wade H, Pan K, Su Q. CREBH: A Complex Array of Regulatory Mechanisms in Nutritional Signaling, Metabolic Inflammation, and Metabolic Disease. Mol Nutr Food Res 2020; 65:e2000771. [DOI: 10.1002/mnfr.202000771] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Henry Wade
- Institute for Global Food Security School of Biological Sciences Queen's University Belfast Belfast BT9 5DL UK
| | - Kaichao Pan
- Institute for Global Food Security School of Biological Sciences Queen's University Belfast Belfast BT9 5DL UK
| | - Qiaozhu Su
- Institute for Global Food Security School of Biological Sciences Queen's University Belfast Belfast BT9 5DL UK
| |
Collapse
|
35
|
Song LY, Wu YZ, Pei XX, Li R, Chen HT, Sun XZ. Pulmonary toxicity and RNA sequencing analyses of mouse in response to exposure to cellulose nanofibrils. Inhal Toxicol 2020; 32:388-401. [PMID: 33043732 DOI: 10.1080/08958378.2020.1831112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The growing applications of nanocelluloses in the fields of advanced nanocomposites, electronics, and medical devices necessitate investigation of their potential adverse effects on human health. The lungs are the primary and the most important route for the entry of nanocelluloses into the human body in occupational settings. However, data on the pulmonary toxicity of cellulose nanofibrils (CNFs) and its molecular mechanism are limited. This study investigated the pulmonary toxicity of CNFs and its genomic expression using the RNA sequencing approach. MATERIALS AND METHODS Female C57BL/6 mice were administered CNFs at 50 μg/mouse by oropharyngeal aspiration. Samples were collected at 3 and 14 days after exposure to CNFs (DAEC). RESULTS At three DAEC, the microscopic sections of lungs revealed a significant inflammatory response. In terms of gene expression alterations, 94 genes were up-regulated, and 107 genes were down-regulated. Most of these differentially expressed genes were involved in the inflammatory and immune responses, including chemokines, NK cells, killer cell lectin-like receptors, CD antigens, T cell-specific GTPases, immunity-related GTPase family M members, and interferon-induced proteins encoding genes. However, only 9 and 26 genes at 14 DAEC were significantly up- and down-regulated, respectively. CONCLUSIONS The pathological analysis of lung sections and the analysis of sequencing data suggested that the homeostasis of mice lungs was restored at 14 DAEC. The findings of this study provide insights into the pulmonary toxicity, and underlying toxicological mechanisms, caused by exposure to CNFs, and are useful for the assessment of the potential toxicity of nanocelluloses.
Collapse
Affiliation(s)
- Li-Ying Song
- College of Engineering, Northeast Agricultural University, Harbin, China
| | - Yun-Zhou Wu
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Xi-Xiang Pei
- Harbin Botai Biological Technology Co., Ltd, Harbin, China
| | - Rui Li
- College of Engineering, Northeast Agricultural University, Harbin, China
| | - Hai-Tao Chen
- College of Engineering, Northeast Agricultural University, Harbin, China
| | - Xiao-Zheng Sun
- College of Engineering, Northeast Agricultural University, Harbin, China
| |
Collapse
|
36
|
Zhang N, Wang Y, Zhang J, Liu B, Deng X, Xin S, Xu K. N-glycosylation of CREBH improves lipid metabolism and attenuates lipotoxicity in NAFLD by modulating PPARα and SCD-1. FASEB J 2020; 34:15338-15363. [PMID: 32996649 DOI: 10.1096/fj.202000836rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cyclic adenosine monophosphate (AMP)-responsive element-binding protein H (CREBH), an endoplasmic reticulum-anchored transcription factor essential for lipid metabolism and inflammation in nonalcoholic fatty liver disease (NAFLD), is covalently modified by N-acetylglucosamine. Glycosylation is a ubiquitous type of protein involved in posttranslational modifications, and plays a critical role in various biological processes. However, the mechanism of glycosylated CREBH remains poorly understood in NAFLD. METHODS CREBH glycosylation mutants were obtained by site-mutation methods. After transfection with plasmids, AML-12, LO2, or HepG2 cells were treated with palmitic acid (PA) proteolysis, tunicamycin (Tm), or their combination. Glycosyltransferase V (GnT-V) was used induce hyperglycosylation to further understand the effect of CREBH. In addition, glycosylation mutant mice and hyperglycosylated mice were generated by lentivirus injection to construct two kinds of NAFLD animal models. The expression of NAFLD-related factors was detected to further verify the role of N-linked glycosylation of CREBH in lipid and sterol metabolism, inflammation, and lipotoxicity. RESULTS N-glycosylation enhanced the ability of CREBH to activate transcription and modulated the production of peroxisome proliferator-activated receptor alpha (PPARα) and stearoyl-CoA desaturase-1 (SCD-1) activity by affecting their promoter-driven transcription activity and protein interactions, leading to reduce lipid deposition and attenuate lipotoxicity. Deglycosylation of CREBH induced by Tm could inhibit the proteolysis of CREBH induced by PA. The addition of unglycosylated CREBH to cells upregulates gene and protein expression of lipogenesis, lipotoxicity, and inflammation, and aggravates liver damage by preventing glycosylation in cells, as well as in mouse models of NAFLD. Furthermore, increased N-glycosylation of CREBH, as achieved by overexpressing GnT-V could significantly improve liver lesion caused by unglycosylation of CREBH. CONCLUSION These findings have important implications for the role of CREBH N-glycosylation in proteolytic activation, and they provide the first link between N-glycosylation of CREBH, lipid metabolism, and lipotoxicity processes in the liver by modulating PPARα and SCD-1. These results provide novel insights into the N-glycosylation of CREBH as a therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Ning Zhang
- Division of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Yuli Wang
- Division of Oncology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Junli Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Beibei Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoling Deng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengliang Xin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Nassir F. Role of acetylation in nonalcoholic fatty liver disease: a focus on SIRT1 and SIRT3. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most prevalent liver chronic disease worldwide. The pathogenesis of NAFLD is complex and involves many metabolic enzymes and multiple pathways. Posttranslational modifications of proteins (PMPs) added another layer of complexity to the pathogenesis of NAFLD. PMPs change protein properties and regulate many biological functions, including cellular localization, stability, intracellular signaling, and protein function. Lysine acetylation is a common reversible PMP that consists of the transfer of an acetyl group from acetyl-coenzyme A (CoA) to a lysine residue on targeted proteins. The deacetylation reaction is catalyzed by deacetylases called sirtuins. This review summarizes the role of acetylation in NAFLD with a focus on sirtuins 1 and 3.
Collapse
Affiliation(s)
- Fatiha Nassir
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
38
|
Borkham-Kamphorst E, Haas U, Van de Leur E, Trevanich A, Weiskirchen R. Chronic Carbon Tetrachloride Applications Induced Hepatocyte Apoptosis in Lipocalin 2 Null Mice Through Endoplasmic Reticulum Stress and Unfolded Protein Response. Int J Mol Sci 2020; 21:ijms21155230. [PMID: 32718038 PMCID: PMC7432394 DOI: 10.3390/ijms21155230] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
The lack of Lipocalin (LCN2) provokes overwhelming endoplasmic reticulum (ER) stress responses in vitro and in acute toxic liver injury models, resulting in hepatocyte apoptosis. LCN2 is an acute phase protein produced in hepatocytes in response to acute liver injuries. In line with these findings we investigated ER stress responses of Lcn2−/− mice in chronic ER stress using a long-term repetitive carbon tetrachloride (CCl4) injection model. We found chronic CCl4 application to enhance ER stress and unfolded protein responses (UPR), including phosphorylation of eukaryotic initiation factor 2α (eIF2α), increased expression of binding immunoglobulin protein (BiP) and glucose-regulated protein 94 (GRP94). IRE1α/TRAF2/JNK signaling enhanced mitochondrial apoptotic pathways, and showed slightly higher in Lcn2−/− mice compared to the wild type counterparts, leading to increased hepatocyte apoptosis well evidenced by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Hepatocyte injuries were confirmed by significant high serum alanine transaminase (ALT) levels in CCl4-treated Lcn2−/− mice. Lcn2−/− mice furthermore developed mild hepatic steatosis, supporting our finding that ER stress promotes lipogenesis. In a previous report we demonstrated that the pharmacological agent tunicamycin (TM) induced ER stress through altered protein glycosylation and induced high amounts of C/EBP-homologous protein (CHOP), resulting in hepatocyte apoptosis. We compared TM-induced ER stress in wild type, Lcn2−/−, and Chop null (Chop−/−) primary hepatocytes and found Chop−/− hepatocytes to attenuate ER stress responses and resist ER stress-induced hepatocyte apoptosis through canonical eIF2α/GADD34 signaling, inhibiting protein synthesis. Unexpectedly, in later stages of TM incubation, Chop−/− hepatocytes resumed activation of IRE1α/JNK/c-Jun and p38/ATF2 signaling, leading to late hepatocyte apoptosis. This interesting observation indicates Chop−/− mice to be unable to absolutely prevent all types of liver injury, while LCN2 protects the hepatocytes by maintaining homeostasis under ER stress conditions.
Collapse
Affiliation(s)
- Erawan Borkham-Kamphorst
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, D-52074 Aachen, Germany; (U.H.); (E.V.d.L.)
- Correspondence: (E.B.-K.); (R.W.); Tel.: +49-241-80-88684 (E.B.-K.); +49-241-80-88683 (R.W.)
| | - Ute Haas
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, D-52074 Aachen, Germany; (U.H.); (E.V.d.L.)
| | - Eddy Van de Leur
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, D-52074 Aachen, Germany; (U.H.); (E.V.d.L.)
| | - Anothai Trevanich
- Department of Statistics, Faculty of Science, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University Hospital, D-52074 Aachen, Germany; (U.H.); (E.V.d.L.)
- Correspondence: (E.B.-K.); (R.W.); Tel.: +49-241-80-88684 (E.B.-K.); +49-241-80-88683 (R.W.)
| |
Collapse
|
39
|
Transcriptional Regulation in Non-Alcoholic Fatty Liver Disease. Metabolites 2020; 10:metabo10070283. [PMID: 32660130 PMCID: PMC7408131 DOI: 10.3390/metabo10070283] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is the primary risk factor for the pathogenesis of non-alcoholic fatty liver disease (NAFLD), the worldwide prevalence of which continues to increase dramatically. The liver plays a pivotal role in the maintenance of whole-body lipid and glucose homeostasis. This is mainly mediated by the transcriptional activation of hepatic pathways that promote glucose and lipid production or utilization in response to the nutritional state of the body. However, in the setting of chronic excessive nutrition, the dysregulation of hepatic transcriptional machinery promotes lipid accumulation, inflammation, metabolic stress, and fibrosis, which culminate in NAFLD. In this review, we provide our current understanding of the transcription factors that have been linked to the pathogenesis and progression of NAFLD. Using publicly available transcriptomic data, we outline the altered activity of transcription factors among humans with NAFLD. By expanding this analysis to common experimental mouse models of NAFLD, we outline the relevance of mouse models to the human pathophysiology at the transcriptional level.
Collapse
|
40
|
Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol 2020; 21:421-438. [PMID: 32457508 DOI: 10.1038/s41580-020-0250-z] [Citation(s) in RCA: 1529] [Impact Index Per Article: 305.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
Abstract
Cellular stress induced by the abnormal accumulation of unfolded or misfolded proteins at the endoplasmic reticulum (ER) is emerging as a possible driver of human diseases, including cancer, diabetes, obesity and neurodegeneration. ER proteostasis surveillance is mediated by the unfolded protein response (UPR), a signal transduction pathway that senses the fidelity of protein folding in the ER lumen. The UPR transmits information about protein folding status to the nucleus and cytosol to adjust the protein folding capacity of the cell or, in the event of chronic damage, induce apoptotic cell death. Recent advances in the understanding of the regulation of UPR signalling and its implications in the pathophysiology of disease might open new therapeutic avenues.
Collapse
Affiliation(s)
- Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,FONDAP Center for Geroscience Brain Health and Metabolism (GERO), Santiago, Chile. .,Program of Cellular and Molecular Biology, Institute of Biomedical Science, University of Chile, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
41
|
Li G, Zhang J, Jiang Q, Liu B, Xu K. CREBH knockout accelerates hepatic fibrosis in mouse models of diet-induced nonalcoholic fatty liver disease. Life Sci 2020; 254:117795. [PMID: 32417373 DOI: 10.1016/j.lfs.2020.117795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/12/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
AIMS The primary focus of this study was to explore the effects of cyclic AMP response element-binding protein H (CREBH) on the development of nonalcoholic fatty liver disease (NAFLD). MATERIALS AND METHODS CREBH knockout (KO) and wildtype (WT) mice were averagely divided into a methionine and choline-deficient (MCD) or high fat (HF) diet group and respective chow diet (CD) groups. Mice were sacrificed after 4-week treatment for MCD model and 24-week treatment for HF model. KEY FINDINGS Characteristics of nonalcoholic steatohepatitis-related liver fibrosis in KO-MCD/HF group were verified by hepatic histological analyses. Compared with WT-MCD/HF group, levels of plasma ALT and hepatic hydroxyproline increased in KO-MCD/HF group. Significantly higher levels of MCP-1, αSMA, Desmin, COL-1, TIMP-1, TGF-β1, TGF-β2 were found while MMP-9 and FGF21 mRNA levels decreased in KO-MCD/HF group. There was also a distinct difference of mRNA levels of TNFα, CTGF and CCND1 in KO-HF group compared with controls. Protein levels of MCP-1, BAX, αSMA, COL-1, TGF-β1 and SMAD2/3 significantly increased in KO-MCD/HF group and CCND1 was also upregulated in KO-HF group compared to their counterparts. SIGNIFICANCE CREBH knockout may primarily regulate the TGF-β1 signaling pathway via TGF-β2 and FGF21 resulting in more severe inflammation and fibrosis in NAFLD.
Collapse
Affiliation(s)
- Guixin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junli Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianqian Jiang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Beibei Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Keshu Xu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
42
|
Saran AR, Dave S, Zarrinpar A. Circadian Rhythms in the Pathogenesis and Treatment of Fatty Liver Disease. Gastroenterology 2020; 158:1948-1966.e1. [PMID: 32061597 PMCID: PMC7279714 DOI: 10.1053/j.gastro.2020.01.050] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
Circadian clock proteins are endogenous timing mechanisms that control the transcription of hundreds of genes. Their integral role in coordinating metabolism has led to their scrutiny in a number of diseases, including nonalcoholic fatty liver disease (NAFLD). Discoordination between central and peripheral circadian rhythms is a core feature of nearly every genetic, dietary, or environmental model of metabolic syndrome and NAFLD. Restricting feeding to a defined daily interval (time-restricted feeding) can synchronize the central and peripheral circadian rhythms, which in turn can prevent or even treat the metabolic syndrome and hepatic steatosis. Importantly, a number of proteins currently under study as drug targets in NAFLD (sterol regulatory element-binding protein [SREBP], acetyl-CoA carboxylase [ACC], peroxisome proliferator-activator receptors [PPARs], and incretins) are modulated by circadian proteins. Thus, the clock can be used to maximize the benefits and minimize the adverse effects of pharmaceutical agents for NAFLD. The circadian clock itself has the potential for use as a target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Anand R. Saran
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA
| | - Shravan Dave
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, California; Veterans Affairs Health Sciences San Diego, La Jolla, California; Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, California; Center for Microbiome Innovation, University of California, San Diego, La Jolla, California.
| |
Collapse
|
43
|
CREBH Improves Diet-Induced Obesity, Insulin Resistance, and Metabolic Disturbances by FGF21-Dependent and FGF21-Independent Mechanisms. iScience 2020; 23:100930. [PMID: 32151974 PMCID: PMC7063134 DOI: 10.1016/j.isci.2020.100930] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/16/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Mice overexpressing the nuclear form of CREBH mainly in the liver (CREBH-Tg) showed suppression of high-fat high-sucrose (HFHS) diet-induced obesity accompanied by an increase in plasma fibroblast growth factor 21 (FGF21) levels. CREBH overexpression induced browning in inguinal white adipose tissue (WAT) and whole-body energy expenditure, which was canceled in Fgf21−/− mice. Deficiency of FGF21 in CREBH-Tg mice mostly canceled the improvement of obesity, but the suppression of inflammation of epidermal WAT, amelioration of insulin resistance, and improvement of glucose metabolism still sustained. Kisspeptin 1 (Kiss1) was identified as a novel hormone target for CREBH to explain these FGF21-independent effects of CREBH. Knockdown of Kiss1 in HFHS-fed CREBH-Tg Fgf21−/− mice showed partially canceled improvement of glucose metabolism. Taken together, we propose that hepatic CREBH pleiotropically improves diet-induced obesity-mediated dysfunctions in peripheral tissues by improving systemic energy metabolism in FGF21-dependent and FGF21-independent mechanisms. Deficiency of FGF21 in CREBH-Tg mice mostly cancels the improvement of obesity CREBH induces browning in iWAT CREBH suppresses inflammation of eWAT CREBH-induced Kiss1 contributes to improvement of glucose metabolism
Collapse
|
44
|
Cebola I. Liver gene regulatory networks: Contributing factors to nonalcoholic fatty liver disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1480. [PMID: 32020788 DOI: 10.1002/wsbm.1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/17/2022]
Abstract
Metabolic diseases such as nonalcoholic fatty liver disease (NAFLD) result from complex interactions between intrinsic and extrinsic factors, including genetics and exposure to obesogenic environments. These risk factors converge in aberrant gene expression patterns in the liver, which are underlined by altered cis-regulatory networks. In homeostasis and in disease states, liver cis-regulatory networks are established by coordinated action of liver-enriched transcription factors (TFs), which define enhancer landscapes, activating broad gene programs with spatiotemporal resolution. Recent advances in DNA sequencing have dramatically expanded our ability to map active transcripts, enhancers and TF cistromes, and to define the 3D chromatin topology that contains these elements. Deployment of these technologies has allowed investigation of the molecular processes that regulate liver development and metabolic homeostasis. Moreover, genomic studies of NAFLD patients and NAFLD models have demonstrated that the liver undergoes pervasive regulatory rewiring in NAFLD, which is reflected by aberrant gene expression profiles. We have therefore achieved an unprecedented level of detail in the understanding of liver cis-regulatory networks, particularly in physiological conditions. Future studies should aim to map active regulatory elements with added levels of resolution, addressing how the chromatin landscapes of different cell lineages contribute to and are altered in NAFLD and NAFLD-associated metabolic states. Such efforts would provide additional clues into the molecular factors that trigger this disease. This article is categorized under: Biological Mechanisms > Metabolism Biological Mechanisms > Regulatory Biology Laboratory Methods and Technologies > Genetic/Genomic Methods.
Collapse
Affiliation(s)
- Inês Cebola
- Department of Metabolism, Digestion and Reproduction, Section of Genetics and Genomics, Imperial College London, London, UK
| |
Collapse
|
45
|
El‐Deen RM, Heeba GH, Abdel‐latif RG, Khalifa MM. Comparative effectiveness of phosphodiesterase 3, 4, and 5 inhibitors in amelioration of high‐fat diet‐induced nonalcoholic fatty liver in rats. Fundam Clin Pharmacol 2020; 34:353-364. [DOI: 10.1111/fcp.12530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Reham M. El‐Deen
- Undersecretary Preventive Sector General Administration Viral Hepatitis Ministry of Health and Population Cairo11516 Egypt
| | - Gehan H. Heeba
- Department of Pharmacology & Toxicology Faculty of Pharmacy Minia University El‐Minia61511 Egypt
| | - Rania G. Abdel‐latif
- Department of Pharmacology & Toxicology Faculty of Pharmacy Minia University El‐Minia61511 Egypt
| | - Mohamed M.A. Khalifa
- Department of Pharmacology & Toxicology Faculty of Pharmacy Minia University El‐Minia61511 Egypt
| |
Collapse
|
46
|
Noh JR, Kim JH, Na SY, Lee IB, Seo YJ, Choi JH, Seo Y, Lee TG, Choi HS, Kim YH, Lee CH. Hepatocyte CREBH deficiency aggravates inflammatory liver injury following chemokine-dependent neutrophil infiltration through upregulation of NF-κB p65 in mice. Arch Toxicol 2019; 94:509-522. [DOI: 10.1007/s00204-019-02633-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/26/2019] [Indexed: 12/31/2022]
|
47
|
Sprooten J, Garg AD. Type I interferons and endoplasmic reticulum stress in health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:63-118. [PMID: 32138904 PMCID: PMC7104985 DOI: 10.1016/bs.ircmb.2019.10.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) comprise of pro-inflammatory cytokines created, as well as sensed, by all nucleated cells with the main objective of blocking pathogens-driven infections. Owing to this broad range of influence, type I IFNs also exhibit critical functions in many sterile inflammatory diseases and immunopathologies, especially those associated with endoplasmic reticulum (ER) stress-driven signaling pathways. Indeed, over the years accumulating evidence has indicated that the presence of ER stress can influence the production, or sensing of, type I IFNs induced by perturbations like pattern recognition receptor (PRR) agonists, infections (bacterial, viral or parasitic) or autoimmunity. In this article we discuss the link between type I IFNs and ER stress in various diseased contexts. We describe how ER stress regulates type I IFNs production or sensing, or how type I IFNs may induce ER stress, in various circumstances like microbial infections, autoimmunity, diabetes, cancer and other ER stress-related contexts.
Collapse
Affiliation(s)
- Jenny Sprooten
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
48
|
Khan HA, Margulies CE. The Role of Mammalian Creb3-Like Transcription Factors in Response to Nutrients. Front Genet 2019; 10:591. [PMID: 31293620 PMCID: PMC6598459 DOI: 10.3389/fgene.2019.00591] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 06/04/2019] [Indexed: 12/18/2022] Open
Abstract
Our ability to overcome the challenges behind metabolic disorders will require a detailed understanding of the regulation of responses to nutrition. The Creb3 transcription factor family appears to have a unique regulatory role that links cellular secretory capacity with development, nutritional state, infection, and other stresses. This role in regulating individual secretory capacity genes could place this family of transcription factors at an important regulatory intersection mediating an animal’s responses to nutrients and other environmental challenges. Interestingly, in both humans and mice, individuals with mutations in Creb3L3/CrebH, one of the Creb3 family members, exhibit hypertriglyceridemia (HTG) thus linking this transcription factor to lipid metabolism. We are beginning to understand how Creb3L3 and related family members are regulated and to dissect the potential redundancy and cross talk between distinct family members, thereby mediating both healthy and pathological responses to the environment. Here, we review the current knowledge on the regulation of Creb3 family transcription factor activity, their target genes, and their role in metabolic disease.
Collapse
Affiliation(s)
- Haris A Khan
- Physiological Chemistry, Biomedical Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Carla E Margulies
- Physiological Chemistry, Biomedical Center, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
49
|
Zhang Q, Zhu Q, Deng R, Zhou F, Zhang L, Wang S, Zhu K, Wang X, Zhou L, Su Q. MS-275 induces hepatic FGF21 expression via H3K18ac-mediated CREBH signal. J Mol Endocrinol 2019; 62:187-196. [PMID: 30893641 DOI: 10.1530/jme-18-0259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022]
Abstract
Fibroblast growth factor 21 (FGF21) plays an important role in the regulation of lipid and glucose metabolism. MS-275, as a class I-specific histone deacetylase (HDAC) inhibitor, has also been reported to affect energy metabolism. In this current study, we investigated the effects of MS-275 on hepatic FGF21 expression in vitro and in vivo and explored whether cAMP-responsive element-binding protein H (CREBH) was involved in the action of MS-275. Our results showed that MS-275 stimulated hepatic FGF21 mRNA and protein expressions in a dose- and time-dependent manner, as well as FGF21 secretion in primary mouse hepatocytes. Serum concentration and hepatic expression of FGF21 were elevated after injection of MS-275, along with increased expressions of genes involved in fatty acid oxidation and ketogenic production (peroxisome proliferator-activated receptor gammacoactivator1α, PGC-1α; carnitine palmitoyl-transferase 1a, CPT1a; 3-hydroxy-3-methylglutaryl-CoA synthase 2, Hmgcs2) as well as improved blood lipid profile. As a proved transcription factor of FGF21, the expression of CREBH was initiated by MS-275, with increased histone H3 lysine 18 acetylation (H3K18ac) signals and hepatocyte nuclear factor 4 alpha (HNF-4α) recruitment in CREBH promoter. Adenovirus-mediated knockdown of CREBH abolished MS-275-induced hepatic FGF21 and lipid metabolism-related gene expressions. These results suggest that MS-275 induces hepatic FGF21 by H3K18ac-mediated CREBH expression.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qin Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruyuan Deng
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Feiye Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Linlin Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shushu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kecheng Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Zhang L, Yao W, Xia J, Wang T, Huang F. Glucagon-Induced Acetylation of Energy-Sensing Factors in Control of Hepatic Metabolism. Int J Mol Sci 2019; 20:ijms20081885. [PMID: 30995792 PMCID: PMC6515121 DOI: 10.3390/ijms20081885] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
The liver is the central organ of glycolipid metabolism, which regulates the metabolism of lipids and glucose to maintain energy homeostasis upon alterations of physiological conditions. Researchers formerly focused on the phosphorylation of glucagon in controlling liver metabolism. Noteworthily, emerging evidence has shown glucagon could additionally induce acetylation to control hepatic metabolism in response to different physiological states. Through inducing acetylation of complex metabolic networks, glucagon interacts extensively with various energy-sensing factors in shifting from glucose metabolism to lipid metabolism during prolonged fasting. In addition, glucagon-induced acetylation of different energy-sensing factors is involved in the advancement of nonalcoholic fatty liver disease (NAFLD) to liver cancer. Here, we summarize the latest findings on glucagon to control hepatic metabolism by inducing acetylation of energy-sensing factors. Finally, we summarize and discuss the potential impact of glucagon on the treatment of liver diseases.
Collapse
Affiliation(s)
- Li Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Weilei Yao
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jun Xia
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Tongxin Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|