1
|
Cartes-Saavedra B, Ghosh A, Hajnóczky G. The roles of mitochondria in global and local intracellular calcium signalling. Nat Rev Mol Cell Biol 2025:10.1038/s41580-024-00820-1. [PMID: 39870977 DOI: 10.1038/s41580-024-00820-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/29/2025]
Abstract
Activation of Ca2+ channels in Ca2+ stores in organelles and the plasma membrane generates cytoplasmic calcium ([Ca2+]c) signals that control almost every aspect of cell function, including metabolism, vesicle fusion and contraction. Mitochondria have a high capacity for Ca2+ uptake and chelation, alongside efficient Ca2+ release mechanisms. Still, mitochondria do not store Ca2+ in a prolonged manner under physiological conditions and lack the capacity to generate global [Ca2+]c signals. However, mitochondria take up Ca2+ at high local [Ca2+]c signals that originate from neighbouring organelles, and also during sustained global elevations of [Ca2+]c. Accumulated Ca2+ in the mitochondria stimulates oxidative metabolism and upon return to the cytoplasm, can produce spatially confined rises in [Ca2+]c to exert control over processes that are sensitive to Ca2+. Thus, the mitochondrial handling of [Ca2+]c is of physiological relevance. Furthermore, dysregulation of mitochondrial Ca2+ handling can contribute to debilitating diseases. We discuss the mechanisms and relevance of mitochondria in local and global calcium signals.
Collapse
Affiliation(s)
- Benjamín Cartes-Saavedra
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Arijita Ghosh
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Ulrich H, Glaser T, Thomas AP. Purinergic signaling in liver disease: calcium signaling and induction of inflammation. Purinergic Signal 2024:10.1007/s11302-024-10044-9. [PMID: 39320433 DOI: 10.1007/s11302-024-10044-9] [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: 02/28/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
Purinergic signaling regulates many metabolic functions and is implicated in liver physiology and pathophysiology. Liver functionality is modulated by ionotropic P2X and metabotropic P2Y receptors, specifically P2Y1, P2Y2, and P2Y6 subtypes, which physiologically exert their influence through calcium signaling, a key second messenger controlling glucose and fat metabolism in hepatocytes. Purinergic receptors, acting through calcium signaling, play an important role in a range of liver diseases. Ionotropic P2X receptors, such as the P2X7 subtype, and certain metabotropic P2Y receptors can induce aberrant intracellular calcium transients that impact normal hepatocyte function and initiate the activation of other liver cell types, including Kupffer and stellate cells. These P2Y- and P2X-dependent intracellular calcium increases are particularly relevant in hepatic disease states, where stellate and Kupffer cells respond with innate immune reactions to challenges, such as excess fat accumulation, chronic alcohol abuse, or infections, and can eventually lead to liver fibrosis. This review explores the consequences of excessive extracellular ATP accumulation, triggering calcium influx through P2X4 and P2X7 receptors, inflammasome activation, and programmed cell death. In addition, P2Y2 receptors contribute to hepatic steatosis and insulin resistance, while inhibiting the expression of P2Y6 receptors can alleviate alcoholic liver steatosis. Adenosine receptors may also contribute to fibrosis through extracellular matrix production by fibroblasts. Thus, pharmacological modulation of P1 and P2 receptors and downstream calcium signaling may open novel therapeutic avenues.
Collapse
Affiliation(s)
- Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Talita Glaser
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA.
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
3
|
Guo M, Liu R, Zhang F, Qu J, Yang Y, Li X. A new perspective on liver diseases: Focusing on the mitochondria-associated endoplasmic reticulum membranes. Pharmacol Res 2024; 208:107409. [PMID: 39284429 DOI: 10.1016/j.phrs.2024.107409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The pathogenesis of liver diseases is multifaceted and intricate, posing a persistent global public health challenge with limited therapeutic options. Therefore, further research into liver diseases is imperative for better comprehension and advancement in treatment strategies. Numerous studies have confirmed the endoplasmic reticulum (ER) and mitochondria as key organelles driving liver diseases. Notably, the mitochondrial-associated ER membranes (MAMs) establish a physical and functional connection between the ER and mitochondria, highlighting the importance of inter-organelle communication in maintaining their functional homeostasis. This review delves into the intricate architecture and regulative mechanism of the integrated MAM that facilitate the physiological transfer of signals and substances between organelles. Additionally, we also provide a detailed overview regarding the varied pathogenic roles of malfunctioning MAM in liver diseases, focusing on its involvement in the progression of ER stress and mitochondrial dysfunction, the regulation of mitochondrial dynamics and Ca2+ transfer, as well as the disruption of lipid and glucose homeostasis. Furthermore, the current challenges and prospects associated with MAM in liver disease research are thoroughly discussed. In conclusion, elucidating the specific structure and function of MAM in different liver diseases may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Mengyu Guo
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 100029, China
| | - Fukun Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China.
| |
Collapse
|
4
|
Parlakgül G, Pang S, Artico LL, Min N, Cagampan E, Villa R, Goncalves RLS, Lee GY, Xu CS, Hotamışlıgil GS, Arruda AP. Spatial mapping of hepatic ER and mitochondria architecture reveals zonated remodeling in fasting and obesity. Nat Commun 2024; 15:3982. [PMID: 38729945 PMCID: PMC11087507 DOI: 10.1038/s41467-024-48272-7] [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: 06/27/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
The hepatocytes within the liver present an immense capacity to adapt to changes in nutrient availability. Here, by using high resolution volume electron microscopy, we map how hepatic subcellular spatial organization is regulated during nutritional fluctuations and as a function of liver zonation. We identify that fasting leads to remodeling of endoplasmic reticulum (ER) architecture in hepatocytes, characterized by the induction of single rough ER sheet around the mitochondria, which becomes larger and flatter. These alterations are enriched in periportal and mid-lobular hepatocytes but not in pericentral hepatocytes. Gain- and loss-of-function in vivo models demonstrate that the Ribosome receptor binding protein1 (RRBP1) is required to enable fasting-induced ER sheet-mitochondria interactions and to regulate hepatic fatty acid oxidation. Endogenous RRBP1 is enriched around periportal and mid-lobular regions of the liver. In obesity, ER-mitochondria interactions are distinct and fasting fails to induce rough ER sheet-mitochondrion interactions. These findings illustrate the importance of a regulated molecular architecture for hepatocyte metabolic flexibility.
Collapse
Affiliation(s)
- Güneş Parlakgül
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Song Pang
- HHMI Janelia Research Campus, Ashburn, VA, USA
- Yale School of Medicine, New Haven, CT, USA
| | - Leonardo L Artico
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Nina Min
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Erika Cagampan
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Reyna Villa
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - Renata L S Goncalves
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Grace Yankun Lee
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - C Shan Xu
- HHMI Janelia Research Campus, Ashburn, VA, USA
- Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Gökhan S Hotamışlıgil
- Department of Molecular Metabolism and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Ana Paula Arruda
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
5
|
Smith-Cortinez N, Heegsma J, Podunavac M, Zakarian A, Cardenas JC, Faber KN. Novel Inositol 1,4,5-Trisphosphate Receptor Inhibitor Antagonizes Hepatic Stellate Cell Activation: A Potential Drug to Treat Liver Fibrosis. Cells 2024; 13:765. [PMID: 38727301 PMCID: PMC11083487 DOI: 10.3390/cells13090765] [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: 02/22/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Liver fibrosis, characterized by excessive extracellular matrix (ECM) deposition, can progress to cirrhosis and increases the risk of liver cancer. Hepatic stellate cells (HSCs) play a pivotal role in fibrosis progression, transitioning from a quiescent to activated state upon liver injury, wherein they proliferate, migrate, and produce ECM. Calcium signaling, involving the inositol 1,4,5-trisphosphate receptor (IP3R), regulates HSC activation. This study investigated the efficacy of a novel IP3R inhibitor, desmethylxestospongin B (dmXeB), in preventing HSC activation. Freshly isolated rat HSCs were activated in vitro in the presence of varying dmXeB concentrations. The dmXeB effectively inhibited HSC proliferation, migration, and expression of fibrosis markers without toxicity to the primary rat hepatocytes or human liver organoids. Furthermore, dmXeB preserved the quiescent phenotype of HSCs marked by retained vitamin A storage. Mechanistically, dmXeB suppressed mitochondrial respiration in activated HSCs while enhancing glycolytic activity. Notably, methyl pyruvate, dimethyl α-ketoglutarate, and nucleoside supplementation all individually restored HSC proliferation despite dmXeB treatment. Overall, dmXeB demonstrates promising anti-fibrotic effects by inhibiting HSC activation via IP3R antagonism without adverse effects on other liver cells. These findings highlight dmXeB as a potential therapeutic agent for liver fibrosis treatment, offering a targeted approach to mitigate liver fibrosis progression and its associated complications.
Collapse
Affiliation(s)
- Natalia Smith-Cortinez
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands
| | - Janette Heegsma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands
| | - Masa Podunavac
- Department of Chemistry and Biochemistry, University of California, Oakland, CA 94607, USA
| | - Armen Zakarian
- Department of Chemistry and Biochemistry, University of California, Oakland, CA 94607, USA
| | - J. César Cardenas
- Department of Chemistry and Biochemistry, University of California, Oakland, CA 94607, USA
- Center for Integrative Biology, Universidad Mayor, Santiago 7510041, Chile
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen (UMCG), 9713 GZ Groningen, The Netherlands
| |
Collapse
|
6
|
Wei J, Liu J, Wang H, Wen K, Ni X, Lin Y, Huang J, You X, Lei Z, Li J, Shen H, Lin Y. Nanoplastic propels diet-induced NAFL to NASH via ER-mitochondrial tether-controlled redox switch. JOURNAL OF HAZARDOUS MATERIALS 2024; 465:133142. [PMID: 38061129 DOI: 10.1016/j.jhazmat.2023.133142] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Nonalcoholic steatohepatitis (NASH) is multifactorial that lifestyle, genetic, and environmental factors contribute to its onset and progression, thereby posing a challenge for therapeutic intervention. Nanoplastic (NP) is emerged as a novel environmental metabolism disruptor but the etiopathogenesis remains largely unknown. In this study, C57BL/6 J mice were fed with normal chow diet (NCD) and high-fat diet (HFD) containing 70 nm polystyrene microspheres (NP). We found that dietary-derived NP adsorbed proteins and agglomerated during the in vivo transportation, enabling diet-induced hepatic steatosis to NASH. Mechanistically, NP promoted liver steatosis by upregulating Fatp2. Furthermore, NP stabilized the Ip3r1, and facilitated ER-mitochondria contacts (MAMs) assembly in the hepatocytes, resulting in mitochondrial Ca2+ overload and redox imbalance. The redox-sensitive Nrf2 was decreased in the liver of NP-exposed mice, which positively regulated miR26a via direct binding to its promoter region [-970 bp to -847 bp and -318 bp to -176 bp]. NP decreased miR26a simultaneously upregulated 10 genes involved in MAMs formation, lipid uptake, inflammation, and fibrosis. Moreover, miR26a inhibition elevated MAMs-tether Vdac1, which promoted the nucleus translocation of NF-κB P65 and Keap1 and functionally inactivated Nrf2, leading to a vicious cycle. Hepatocyte-specific overexpressing miR26a effectively restored ER-mitochondria miscommunication and ameliorated NASH phenotype in NP-exposed and Keap1-overexpressed mice on HFD. The hepatic MAM-tethers/Nrf2/miR26a feedback loop is an essential metabolic switch from simple steatosis to NASH and a promising therapeutic target for oxidative stress-associated liver damage and NASH.
Collapse
Affiliation(s)
- Jie Wei
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jintao Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Huan Wang
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Kai Wen
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Xiuye Ni
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Yilong Lin
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Jingru Huang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Xiang You
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Zhao Lei
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China
| | - Juan Li
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Heqing Shen
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China.
| | - Yi Lin
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, China.
| |
Collapse
|
7
|
Albalawi SS, Aljabri A, Alshibani M, Al-Gayyar MM. The Involvement of Calcium Channels in the Endoplasmic Reticulum Membrane in Nonalcoholic Fatty Liver Disease Pathogenesis. Cureus 2023; 15:e49150. [PMID: 38024063 PMCID: PMC10663096 DOI: 10.7759/cureus.49150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent and complex condition that affects millions of people globally. It occurs when fat, primarily triglycerides, accumulates in liver cells, leading to inflammation and damage. Calcium, an essential mineral, is involved in various physiological processes, including the regeneration process following liver injury. The endoplasmic reticulum (ER), a complex organelle involved in protein synthesis and lipid metabolism, regulates intracellular calcium levels. Dysregulation of this process can lead to calcium overload, oxidative stress, and cellular damage, all of which are hallmarks of NAFLD. Inositol 1,4,5-trisphosphate receptor (IP3R), a type of calcium ion channel, is found throughout the body, including the liver. IP3R is classified into three subtypes: IP3R1, IP3R2, and IP3R3, and it plays a critical role in regulating intracellular calcium levels. However, excessive calcium accumulation in the mitochondria due to an overload of calcium ions or increased IP3R activity can lead to NAFLD. Therefore, targeting calcium channels in the ER membrane may represent a promising therapeutic strategy for preventing and treating this increasingly prevalent metabolic disorder. It may help prevent mitochondrial calcium accumulation and reduce the risk of hepatic damage. This review article aimed to review the relationship between IP3R modulation and the pathogenicity of NAFLD, providing valuable insights to help researchers develop more effective treatments for the condition.
Collapse
Affiliation(s)
- Sarah S Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | - Ahmed Aljabri
- Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, SAU
- Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | - Mohannad Alshibani
- Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, SAU
| | - Mohammed M Al-Gayyar
- Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
- Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, EGY
| |
Collapse
|
8
|
Song H, Zhang X, Wang J, Wu Y, Xiong T, Shen J, Lin R, Xiao T, Lin W. The regulatory role of adipocyte mitochondrial homeostasis in metabolism-related diseases. Front Physiol 2023; 14:1261204. [PMID: 37920803 PMCID: PMC10619862 DOI: 10.3389/fphys.2023.1261204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/09/2023] [Indexed: 11/04/2023] Open
Abstract
Adipose tissue is the most important energy storage organ in the body, maintaining its normal energy metabolism function and playing a vital role in keeping the energy balance of the body to avoid the harm caused by obesity and a series of related diseases resulting from abnormal energy metabolism. The dysfunction of adipose tissue is closely related to the occurrence of diseases related to obesity metabolism. Among various organelles, mitochondria are the main site of energy metabolism, and mitochondria maintain their quality through autophagy, biogenesis, transfer, and dynamics, which play an important role in maintaining metabolic homeostasis of adipocytes. On the other hand, mitochondria have mitochondrial genomes which are vulnerable to damage due to the lack of protective structures and their proximity to sites of reactive oxygen species generation, thus affecting mitochondrial function. Notably, mitochondria are closely related to other organelles in adipocytes, such as lipid droplets and the endoplasmic reticulum, which enhances the function of mitochondria and other organelles and regulates energy metabolism processes, thus reducing the occurrence of obesity-related diseases. This article introduces the structure and quality control of mitochondria in adipocytes and their interactions with other organelles in adipocytes, aiming to provide a new perspective on the regulation of mitochondrial homeostasis in adipocytes on the occurrence of obesity-related diseases, and to provide theoretical reference for further revealing the molecular mechanism of mitochondrial homeostasis in adipocytes on the occurrence of obesity-related diseases.
Collapse
Affiliation(s)
- Hongbing Song
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Xiaohan Zhang
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Jing Wang
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Yanling Wu
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Taimin Xiong
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Jieqiong Shen
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Ruiyi Lin
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Tianfang Xiao
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Weimin Lin
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Zhang J, Zheng Y, Martens L, Pfeiffer AFH. The Regulation and Secretion of Glucagon in Response to Nutrient Composition: Unraveling Their Intricate Mechanisms. Nutrients 2023; 15:3913. [PMID: 37764697 PMCID: PMC10536047 DOI: 10.3390/nu15183913] [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: 08/01/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Glucagon was initially regarded as a hyperglycemic substance; however, recent research has revealed its broader role in metabolism, encompassing effects on glucose, amino acids (AAs), and lipid metabolism. Notably, the interplay of glucagon with nutrient intake, particularly of AAs, and non-nutrient components is central to its secretion. Fasting and postprandial hyperglucagonemia have long been linked to the development and progression of type 2 diabetes (T2DM). However, recent studies have brought to light the positive impact of glucagon agonists on lipid metabolism and energy homeostasis. This review explores the multifaceted actions of glucagon, focusing on its regulation, signaling pathways, and effects on glucose, AAs, and lipid metabolism. The interplay between glucagon and other hormones, including insulin and incretins, is examined to provide a mechanistic understanding of its functions. Notably, the liver-α-cell axis, which involves glucagon and amino acids, emerges as a critical aspect of metabolic regulation. The dysregulation of glucagon secretion and its impact on conditions such as T2DM are discussed. The review highlights the potential therapeutic applications of targeting the glucagon pathway in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jiudan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| | - Yang Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Lisa Martens
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
- Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| |
Collapse
|
10
|
Humbert A, Lefebvre R, Nawrot M, Caussy C, Rieusset J. Calcium signalling in hepatic metabolism: Health and diseases. Cell Calcium 2023; 114:102780. [PMID: 37506596 DOI: 10.1016/j.ceca.2023.102780] [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: 02/28/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023]
Abstract
The flexibility between the wide array of hepatic functions relies on calcium (Ca2+) signalling. Indeed, Ca2+ is implicated in the control of many intracellular functions as well as intercellular communication. Thus, hepatocytes adapt their Ca2+ signalling depending on their nutritional and hormonal environment, leading to opposite cellular functions, such as glucose storage or synthesis. Interestingly, hepatic metabolic diseases, such as obesity, type 2 diabetes and non-alcoholic fatty liver diseases, are associated with impaired Ca2+ signalling. Here, we present the hepatocytes' toolkit for Ca2+ signalling, complete with regulation systems and signalling pathways activated by nutrients and hormones. We further discuss the current knowledge on the molecular mechanisms leading to alterations of Ca2+ signalling in hepatic metabolic diseases, and review the literature on the clinical impact of Ca2+-targeting therapeutics.
Collapse
Affiliation(s)
- Alexandre Humbert
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Rémy Lefebvre
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Margaux Nawrot
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France
| | - Cyrielle Caussy
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France; Département Endocrinologie, Diabète et Nutrition, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Jennifer Rieusset
- Laboratoire CarMeN, INSERM U-1060, INRAE U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre-Bénite, France.
| |
Collapse
|
11
|
Thoudam T, Chanda D, Lee JY, Jung MK, Sinam IS, Kim BG, Park BY, Kwon WH, Kim HJ, Kim M, Lim CW, Lee H, Huh YH, Miller CA, Saxena R, Skill NJ, Huda N, Kusumanchi P, Ma J, Yang Z, Kim MJ, Mun JY, Harris RA, Jeon JH, Liangpunsakul S, Lee IK. Enhanced Ca 2+-channeling complex formation at the ER-mitochondria interface underlies the pathogenesis of alcohol-associated liver disease. Nat Commun 2023; 14:1703. [PMID: 36973273 PMCID: PMC10042999 DOI: 10.1038/s41467-023-37214-4] [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: 03/22/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Abstract
Ca2+ overload-induced mitochondrial dysfunction is considered as a major contributing factor in the pathogenesis of alcohol-associated liver disease (ALD). However, the initiating factors that drive mitochondrial Ca2+ accumulation in ALD remain elusive. Here, we demonstrate that an aberrant increase in hepatic GRP75-mediated mitochondria-associated ER membrane (MAM) Ca2+-channeling (MCC) complex formation promotes mitochondrial dysfunction in vitro and in male mouse model of ALD. Unbiased transcriptomic analysis reveals PDK4 as a prominently inducible MAM kinase in ALD. Analysis of human ALD cohorts further corroborate these findings. Additional mass spectrometry analysis unveils GRP75 as a downstream phosphorylation target of PDK4. Conversely, non-phosphorylatable GRP75 mutation or genetic ablation of PDK4 prevents alcohol-induced MCC complex formation and subsequent mitochondrial Ca2+ accumulation and dysfunction. Finally, ectopic induction of MAM formation reverses the protective effect of PDK4 deficiency in alcohol-induced liver injury. Together, our study defines a mediatory role of PDK4 in promoting mitochondrial dysfunction in ALD.
Collapse
Affiliation(s)
- Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Dipanjan Chanda
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jung Yi Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Min-Kyo Jung
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ibotombi Singh Sinam
- Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Byung-Gyu Kim
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, Republic of Korea
| | - Bo-Yoon Park
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Woong Hee Kwon
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hyo-Jeong Kim
- Electron Microscopy Research Center, Korea Basic Science Institute, Ochang, Chungbuk, Republic of Korea
| | - Myeongjin Kim
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Chae Won Lim
- Bio-Medical Research Institute, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Hoyul Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Ochang, Chungbuk, Republic of Korea
| | - Caroline A Miller
- Electron Microscopy Core, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas J Skill
- Department of Surgery, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Nazmul Huda
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Praveen Kusumanchi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jing Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Min-Ji Kim
- Department of Internal Medicine, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Ji Young Mun
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jae-Han Jeon
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea.
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
| |
Collapse
|
12
|
Arruda AP, Parlakgül G. Endoplasmic Reticulum Architecture and Inter-Organelle Communication in Metabolic Health and Disease. Cold Spring Harb Perspect Biol 2023; 15:a041261. [PMID: 35940911 PMCID: PMC9899651 DOI: 10.1101/cshperspect.a041261] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The endoplasmic reticulum (ER) is a key organelle involved in the regulation of lipid and glucose metabolism, proteostasis, Ca2+ signaling, and detoxification. The structural organization of the ER is very dynamic and complex, with distinct subdomains such as the nuclear envelope and the peripheral ER organized into ER sheets and tubules. ER also forms physical contact sites with all other cellular organelles and with the plasma membrane. Both form and function of the ER are highly adaptive, with a potent capacity to respond to transient changes in environmental cues such as nutritional fluctuations. However, under obesity-induced chronic stress, the ER fails to adapt, leading to ER dysfunction and the development of metabolic pathologies such as insulin resistance and fatty liver disease. Here, we discuss how the remodeling of ER structure and contact sites with other organelles results in diversification of metabolic function and how perturbations to this structural flexibility by chronic overnutrition contribute to ER dysfunction and metabolic pathologies in obesity.
Collapse
Affiliation(s)
- Ana Paula Arruda
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA
- Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| | - Güneş Parlakgül
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, California 94720, USA
- Sabri Ülker Center for Metabolic Research and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
13
|
Ismail M, Zhang X, Taha R, Elhafiz M, Zhang Q, Yousef BA, Huang X, Jiang Z, Zhang L, Sun L. Expression profiles of lncRNAs and their possible regulatory role in monocrotaline-induced HSOS in rats. Front Genet 2023; 14:1041266. [PMID: 36777738 PMCID: PMC9909345 DOI: 10.3389/fgene.2023.1041266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Aims: Long non-coding RNAs (lncRNAs) contribute to the regulation of vital physiological processes and play a role in the pathogenesis of many diseases. Monocrotaline (MCT) can cause large-scale outbreaks of toxic liver disease in humans and animals in the form of hepatic sinusoidal obstruction syndrome (HSOS). Although many experiments have been carried out to explain the pathogenesis of Monocrotaline-induced hepatic sinusoidal obstruction syndrome and to develop treatments for it, no studies have examined the role of Long non-coding RNAs in this condition. This study aimed to investigate the Long non-coding RNAs-mRNA regulation network in Monocrotaline-induced hepatic sinusoidal obstruction syndrome in rats. Main methods: We established a model for MCT-induced hepatic sinusoidal obstruction syndrome, and then carried out microarray for liver tissues of SD rats in a model of early hepatic sinusoidal obstruction syndrome (12 h Monocrotaline treatment vs. control group) to investigate the differentially expressed Long non-coding RNAs and mRNAs in early hepatotoxicity. This was followed by RT-PCR analysis of selected Long non-coding RNAs, which were markedly altered. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genome analyses were also conducted. Key findings: 176 Long non-coding RNAs (63 downregulated and 113 upregulated) and 4,221 mRNAs (2,385 downregulated and 1836 upregulated) were differentially expressed in the Monocrotaline-treated group compared to the control group. The biological processes identified in GO enrichment analysis as playing a role in hepatotoxicity were positive regulation of guanosine triphosphate phosphohydrolase, liver development, and the oxidation-reduction process. Pathway analysis revealed that the metabolism pathways, gap junction, and ribosome biogenesis in eukaryotes were closely related to Monocrotaline-induced hepatotoxicity. According to these analyses, LOC102552718 might play an essential role in hepatotoxicity mechanisms by regulating the expression of inositol 1,4,5-trisphosphate receptor-1 (Itpr-1). Significance: This study provides a basis for further research on the molecular mechanisms underlying Monocrotaline-induced hepatotoxicity and its treatment, especially in the early stage, when successful treatment is critical before irreversible liver damage occurs.
Collapse
Affiliation(s)
- Mohammed Ismail
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China,Department of Pharmacology, Faculty of Medicine and health science, Dongola University, Dongola, Sudan
| | - Xi Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Reham Taha
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Muhanad Elhafiz
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Qianwen Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Bashir A. Yousef
- Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Xin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Zhenzhou Jiang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China,Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China,*Correspondence: Luyong Zhang, ; Lixin Sun,
| | - Lixin Sun
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing, China,*Correspondence: Luyong Zhang, ; Lixin Sun,
| |
Collapse
|
14
|
Duponchel S, Monnier L, Molle J, Bendridi N, Alam MR, Gaballah A, Grigorov B, Ivanov A, Schmiel M, Odenthal M, Ovize M, Rieusset J, Zoulim F, Bartosch B. Hepatitis C virus replication requires integrity of mitochondria-associated ER membranes. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2022; 5:100647. [PMID: 36718430 PMCID: PMC9883273 DOI: 10.1016/j.jhepr.2022.100647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022]
Abstract
Background & Aims Chronic HCV infection causes cellular stress, fibrosis and predisposes to hepatocarcinogenesis. Mitochondria play key roles in orchestrating stress responses by regulating bioenergetics, inflammation and apoptosis. To better understand the role of mitochondria in the viral life cycle and disease progression of chronic hepatitis C, we studied morphological and functional mitochondrial alterations induced by HCV using productively infected hepatoma cells and patient livers. Methods Biochemical and imaging assays were used to assess localization of cellular and viral proteins and mitochondrial functions in cell cultures and liver biopsies. Cyclophilin D (CypD) knockout was performed using CRISPR/Cas9 technology. Viral replication was quantified by quantitative reverse-transcription PCR and western blotting. Results Several HCV proteins were found to associate with mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs), the points of contact between the ER and mitochondria. Downregulation of CypD, which is known to disrupt MAM integrity, reduced viral replication, suggesting that MAMs play an important role in the viral life cycle. This process was rescued by ectopic CypD expression. Furthermore, HCV proteins were found to associate with voltage dependent anion channel 1 (VDAC1) at MAMs and to reduce VDAC1 protein levels at MAMs in vitro and in patient biopsies. This association did not affect MAM-associated functions in glucose homeostasis and Ca2+ signaling. Conclusions HCV proteins associate specifically with MAMs and MAMs play an important role in viral replication. The association between viral proteins and MAMs did not impact Ca2+ signaling between the ER and mitochondria or glucose homeostasis. Whether additional functions of MAMs and/or VDAC are impacted by HCV and contribute to the associated pathology remains to be assessed. Impact and implications Hepatitis C virus infects the liver, where it causes inflammation, cell damage and increases the long-term risk of liver cancer. We show that several HCV proteins interact with mitochondria in liver cells and alter the composition of mitochondrial subdomains. Importantly, HCV requires the architecture of these mitochondrial subdomains to remain intact for efficient viral replication.
Collapse
Key Words
- CypD, cyclophilin D
- DMVs, double membrane vesicles
- EM, electron microscopy
- ER, endoplasmic reticulum
- Grp75, glucose-regulated protein 75
- HCC, hepatocellular carcinoma
- HCVcc, cell culture-derived HCV
- IP, immunoprecipitation
- IP3R1, inositol trisphosphate receptor 1
- KO, knockout
- MAMs, mitochondria-associated ER membranes
- MOI, multiplicity of infection
- OMM, outer mitochondrial membrane
- PLA, proximity ligation assay
- S1R, sigma 1 receptor
- VDAC, voltage-dependent anion channel
- dpi, days post infection
- fibrosis
- hepatitis C virus
- mitochondria-associated ER membranes
- voltage-dependent anion channel 1
Collapse
Affiliation(s)
- Sarah Duponchel
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Lea Monnier
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Jennifer Molle
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Nadia Bendridi
- Laboratoire CarMeN, INSERM U-1060, INRA U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre Bénite, 69495, France
| | - Muhammad Rizwan Alam
- CarMeN Laboratory, Hôpital Louis Pradel, Hospices Civils de Lyon, Université de Lyon and Explorations Fonctionnelles Cardiovasculaires, INSERM U1060, Lyon, France
| | - Ahmed Gaballah
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France,Microbiology Department, Medical Research Institute, Alexandria University, Egypt
| | - Boyan Grigorov
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France
| | - Alexander Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marcel Schmiel
- Institute of Pathology, University Hospital of Cologne and Center for Molecular Medicine (CMMC), University of Cologne, Germany
| | - Margarete Odenthal
- Institute of Pathology, University Hospital of Cologne and Center for Molecular Medicine (CMMC), University of Cologne, Germany
| | - Michel Ovize
- CarMeN Laboratory, Hôpital Louis Pradel, Hospices Civils de Lyon, Université de Lyon and Explorations Fonctionnelles Cardiovasculaires, INSERM U1060, Lyon, France
| | - Jennifer Rieusset
- Laboratoire CarMeN, INSERM U-1060, INRA U-1397, Université Lyon, Université Claude Bernard Lyon 1, Pierre Bénite, 69495, France
| | - Fabien Zoulim
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France,Hospices Civils de Lyon, France
| | - Birke Bartosch
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, 69434, France,Corresponding author. Address: Cancer Research Center Lyon, 151 cours Albert Thomas, 69434 Lyon, France; Tel.: 0033472681975, fax: 0033472681971
| |
Collapse
|
15
|
Zheng W, Sun Q, Li L, Cheng Y, Chen Y, Lv M, Xiang X. Role of endoplasmic reticulum stress in hepatic glucose and lipid metabolism and therapeutic strategies for metabolic liver disease. Int Immunopharmacol 2022; 113:109458. [DOI: 10.1016/j.intimp.2022.109458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/22/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022]
|
16
|
Perry RJ. Regulation of Hepatic Lipid and Glucose Metabolism by INSP3R1. Diabetes 2022; 71:1834-1841. [PMID: 35657697 PMCID: PMC9450566 DOI: 10.2337/dbi22-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022]
Abstract
With the rising epidemics of obesity and nonalcoholic fatty liver disease (NAFLD) and its downstream consequences including steatohepatitis, cirrhosis, and type 2 diabetes in the U.S. and worldwide, new therapeutic approaches are urgently needed to treat these devastating conditions. Glucagon, known for a century to be a glucose-raising hormone and clearly demonstrated to contribute to fasting and postprandial hyperglycemia in both type 1 and type 2 diabetes, represents an unlikely target to improve health in those with metabolic syndrome. However, recent work from our group and others' identifies an unexpected role for glucagon as a potential means of treating NAFLD, improving insulin sensitivity, and improving the lipid profile. We propose a unifying, calcium-dependent mechanism for glucagon's effects both to stimulate hepatic gluconeogenesis and to enhance hepatic mitochondrial oxidation: signaling through the inositol 1,4,5-trisphosphate receptor type 1 (INSP3R1), glucagon activates phospholipase C (PKC)/protein kinase A (PKA) signaling to enhance adipose triglyceride lipase (ATGL)-dependent intrahepatic lipolysis and, in turn, increase cytosolic gluconeogenesis by allosteric activation of pyruvate carboxylase. Simultaneously in the mitochondria, calcium transferred through mitochondria-associated membranes activates several dehydrogenases in the tricarboxylic acid cycle, correlated with an increase in mitochondrial energy expenditure and reduction in ectopic lipid. This model suggests that short-term, cyclic treatment with glucagon or other INSP3R1 antagonists could hold promise as a means to reset lipid homeostasis in patients with NAFLD.
Collapse
Affiliation(s)
- Rachel J. Perry
- Section of Endocrinology & Metabolism, Department of Internal Medicine, and Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
17
|
Jin C, Felli E, Lange NF, Berzigotti A, Gracia-Sancho J, Dufour JF. Endoplasmic Reticulum and Mitochondria Contacts Correlate with the Presence and Severity of NASH in Humans. Int J Mol Sci 2022; 23:ijms23158348. [PMID: 35955482 PMCID: PMC9369179 DOI: 10.3390/ijms23158348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
The interaction between the mitochondria and the endoplasmic reticulum (ER) is essential for hepatocyte function. An increase in ER–mitochondria contacts (ERMCs) is associated with various metabolic diseases. Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and type 2 diabetes, and its progressive form non-alcoholic steatohepatitis (NASH) can lead to cirrhosis and hepatocellular carcinoma. However, the role of ERMCs in the progression of NAFL to NASH is still unclear. We assessed whether ERMCs could correlate with NAFLD severity. We used a proximity ligation assay to measure the abundance of ERMCs in liver biopsies from patients with biopsy-proven NAFLD (n = 48) and correlated the results with histological and metabolic syndrome (MetS) features. NAFLD patients were included according to inclusion and exclusion criteria, and then assigned to NAFL (n = 9) and NASH (n = 39) groups. ERMCs density could discriminate NASH from NAFL (sensitivity 61.5%, specificity 100%). ERMCs abundance correlated with hepatocellular ballooning. Moreover, the density of ERMCs increased with an increase in the number of MetS features. In conclusion, ERMCs increased from NAFL to NASH, in parallel with the number of MetS features, supporting a role for this interaction in the pathophysiology of NASH.
Collapse
Affiliation(s)
- Chaonan Jin
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland; (C.J.); (E.F.); (N.F.L.); (A.B.)
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland; (C.J.); (E.F.); (N.F.L.); (A.B.)
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
| | - Naomi Franziska Lange
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland; (C.J.); (E.F.); (N.F.L.); (A.B.)
- Graduate School for Health Sciences, University of Bern, 3012 Bern, Switzerland
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland; (C.J.); (E.F.); (N.F.L.); (A.B.)
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3012 Bern, Switzerland; (C.J.); (E.F.); (N.F.L.); (A.B.)
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
- Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, 08036 Barcelona, Spain
- Correspondence: (J.G.-S.); (J.-F.D.)
| | - Jean-François Dufour
- Department for BioMedical Research, Hepatology, University of Bern, 3012 Bern, Switzerland
- Correspondence: (J.G.-S.); (J.-F.D.)
| |
Collapse
|
18
|
Chen X, Zhang L, Zheng L, Tuo B. Role of Ca 2+ channels in non-alcoholic fatty liver disease and their implications for therapeutic strategies (Review). Int J Mol Med 2022; 50:113. [PMID: 35796003 PMCID: PMC9282635 DOI: 10.3892/ijmm.2022.5169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/07/2022] [Indexed: 01/10/2023] Open
Affiliation(s)
- Xingyue Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Li Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Liming Zheng
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
19
|
Zhao J, Zhang H, Fan X, Yu X, Huai J. Lipid Dyshomeostasis and Inherited Cerebellar Ataxia. Mol Neurobiol 2022; 59:3800-3828. [PMID: 35420383 PMCID: PMC9148275 DOI: 10.1007/s12035-022-02826-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/01/2022] [Indexed: 12/04/2022]
Abstract
Cerebellar ataxia is a form of ataxia that originates from dysfunction of the cerebellum, but may involve additional neurological tissues. Its clinical symptoms are mainly characterized by the absence of voluntary muscle coordination and loss of control of movement with varying manifestations due to differences in severity, in the site of cerebellar damage and in the involvement of extracerebellar tissues. Cerebellar ataxia may be sporadic, acquired, and hereditary. Hereditary ataxia accounts for the majority of cases. Hereditary ataxia has been tentatively divided into several subtypes by scientists in the field, and nearly all of them remain incurable. This is mainly because the detailed mechanisms of these cerebellar disorders are incompletely understood. To precisely diagnose and treat these diseases, studies on their molecular mechanisms have been conducted extensively in the past. Accumulating evidence has demonstrated that some common pathogenic mechanisms exist within each subtype of inherited ataxia. However, no reports have indicated whether there is a common mechanism among the different subtypes of inherited cerebellar ataxia. In this review, we summarize the available references and databases on neurological disorders characterized by cerebellar ataxia and show that a subset of genes involved in lipid homeostasis form a new group that may cause ataxic disorders through a common mechanism. This common signaling pathway can provide a valuable reference for future diagnosis and treatment of ataxic disorders.
Collapse
Affiliation(s)
- Jin Zhao
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Huan Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xueyu Fan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xue Yu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jisen Huai
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Xinxiang, 453000, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
20
|
Han B, Zhen F, Zheng XS, Hu J, Chen XS. Systematic analysis of the expression and prognostic value of ITPR1 and correlation with tumor infiltrating immune cells in breast cancer. BMC Cancer 2022; 22:297. [PMID: 35313846 PMCID: PMC8939201 DOI: 10.1186/s12885-022-09410-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND ITPR1 is a key gene for autophagy, but its biological function is still unclear, and there are few studies on the correlation between ITPR1 gene expression and the occurrence and development of breast cancer. METHODS Analyze the expression of ITPR1 through online databases such as Oncomine and TIMER. Kaplan-Meier plotter and other databases were used to evaluate the impact of ITPR1 on clinical prognosis. The expression of ITPR1 in analysis of 145 cases of breast cancer and 30 cases of adjacent normal tissue was detected by Immunohistochemistry. Statistical analysis was used to evaluate the clinical relevance and prognostic significance of abnormally expressed proteins. And the Western Blot was used to detect the expression of ITPR1 between breast cancer tissues and cells. The TIMER database studied the relationship between ITPR1 and cancer immune infiltration. And used the ROC plotter database to predict the response of ITPR1 to chemotherapy, endocrine therapy and anti-HER2 therapy in patients with breast cancer. RESULTS Compared with normal breast samples, ITPR1 was significantly lower in patients with breast cancer. And the increased expression of ITPR1 mRNA was closely related to longer overall survival (OS), distant metastasis free survival (DMFS), disease specific survival (DSS) and relapse free survival (RFS) in breast cancer. And the expression level of ITPR1 was higher in patients treated with chemotherapy than untreated patients. In addition, the expression of ITPR1 was positively correlated with related gene markers of immune cells in different types of breast cancer, especially with BRCA basal tissue breast cancer. CONCLUSION ITPR1 was lower expressed in breast cancer. The higher expression of ITPR1 suggested favorable prognosis for patients. ITPR1 was related to the level of immune infiltration, especially in BRCA-Basal patients. All research results indicated that ITPR1 might affect breast cancer prognosis and participate in immune regulation. In short, ITPR1 might be a potential target for breast cancer therapy.
Collapse
Affiliation(s)
- Bing Han
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Fang Zhen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Xiu-Shuang Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, 150001, China
| | - Jing Hu
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| | - Xue-Song Chen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
21
|
Yin H, Shi A, Wu J. Platelet-Activating Factor Promotes the Development of Non-Alcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2022; 15:2003-2030. [PMID: 35837578 PMCID: PMC9275506 DOI: 10.2147/dmso.s367483] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifaceted clinicopathological syndrome characterised by excessive hepatic lipid accumulation that causes steatosis, excluding alcoholic factors. Platelet-activating factor (PAF), a biologically active lipid transmitter, induces platelet activation upon binding to the PAF receptor. Recent studies have found that PAF is associated with gamma-glutamyl transferase, which is an indicator of liver disease. Moreover, PAF can stimulate hepatic lipid synthesis and cause hypertriglyceridaemia. Furthermore, the knockdown of the PAF receptor gene in the animal models of NAFLD helped reduce the inflammatory response, improve glucose homeostasis and delay the development of NAFLD. These findings suggest that PAF is associated with NAFLD development. According to reports, patients with NAFLD or animal models have marked platelet activation abnormalities, mainly manifested as enhanced platelet adhesion and aggregation and altered blood rheology. Pharmacological interventions were accompanied by remission of abnormal platelet activation and significant improvement in liver function and lipids in the animal model of NAFLD. These confirm that platelet activation may accompany a critical importance in NAFLD development and progression. However, how PAFs are involved in the NAFLD signalling pathway needs further investigation. In this paper, we review the relevant literature in recent years and discuss the role played by PAF in NAFLD development. It is important to elucidate the pathogenesis of NAFLD and to find effective interventions for treatment.
Collapse
Affiliation(s)
- Hang Yin
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Anhua Shi
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
| | - Junzi Wu
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China
- Correspondence: Junzi Wu; Anhua Shi, Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China, Tel/Fax +86 187 8855 7524; +86 138 8885 0813, Email ;
| |
Collapse
|
22
|
Chen CC, Hsu LW, Chen KD, Chiu KW, Chen CL, Huang KT. Emerging Roles of Calcium Signaling in the Development of Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2021; 23:ijms23010256. [PMID: 35008682 PMCID: PMC8745268 DOI: 10.3390/ijms23010256] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 01/10/2023] Open
Abstract
The liver plays a central role in energy metabolism. Dysregulated hepatic lipid metabolism is a major cause of non-alcoholic fatty liver disease (NAFLD), a chronic liver disorder closely linked to obesity and insulin resistance. NAFLD is rapidly emerging as a global health problem with currently no approved therapy. While early stages of NAFLD are often considered benign, the disease can progress to an advanced stage that involves chronic inflammation, with increased risk for developing end-stage disease including fibrosis and liver cancer. Hence, there is an urgent need to identify potential pharmacological targets. Ca2+ is an essential signaling molecule involved in a myriad of cellular processes. Intracellular Ca2+ is intricately compartmentalized, and the Ca2+ flow is tightly controlled by a network of Ca2+ transport and buffering proteins. Impaired Ca2+ signaling is strongly associated with endoplasmic reticulum stress, mitochondrial dysfunction and autophagic defects, all of which are etiological factors of NAFLD. In this review, we describe the recent advances that underscore the critical role of dysregulated Ca2+ homeostasis in lipid metabolic abnormalities and discuss the feasibility of targeting Ca2+ signaling as a potential therapeutic approach.
Collapse
Affiliation(s)
- Chien-Chih Chen
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Li-Wen Hsu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
| | - Kuang-Den Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - King-Wah Chiu
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Division of Hepato-Gastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Chao-Long Chen
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
| | - Kuang-Tzu Huang
- Liver Transplantation Center, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan; (L.-W.H.); (K.-D.C.); (K.-W.C.); (C.-L.C.)
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- Correspondence: ; Tel.: +886-7-731-7123 (ext. 8193)
| |
Collapse
|
23
|
Ye L, Zeng Q, Ling M, Ma R, Chen H, Lin F, Li Z, Pan L. Inhibition of IP3R/Ca2+ Dysregulation Protects Mice From Ventilator-Induced Lung Injury via Endoplasmic Reticulum and Mitochondrial Pathways. Front Immunol 2021; 12:729094. [PMID: 34603302 PMCID: PMC8479188 DOI: 10.3389/fimmu.2021.729094] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/31/2021] [Indexed: 01/10/2023] Open
Abstract
Rationale Disruption of intracellular calcium (Ca2+) homeostasis is implicated in inflammatory responses. Here we investigated endoplasmic reticulum (ER) Ca2+ efflux through the Inositol 1,4,5-trisphosphate receptor (IP3R) as a potential mechanism of inflammatory pathophysiology in a ventilator-induced lung injury (VILI) mouse model. Methods C57BL/6 mice were exposed to mechanical ventilation using high tidal volume (HTV). Mice were pretreated with the IP3R agonist carbachol, IP3R inhibitor 2-aminoethoxydiphenyl borate (2-APB) or the Ca2+ chelator BAPTA-AM. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected to measure Ca2+ concentrations, inflammatory responses and mRNA/protein expression associated with ER stress, NLRP3 inflammasome activation and inflammation. Analyses were conducted in concert with cultured murine lung cell lines. Results Lungs from mice subjected to HTV displayed upregulated IP3R expression in ER and mitochondrial-associated-membranes (MAMs), with enhanced formation of MAMs. Moreover, HTV disrupted Ca2+ homeostasis, with increased flux from the ER to the cytoplasm and mitochondria. Administration of carbachol aggravated HTV-induced lung injury and inflammation while pretreatment with 2-APB or BAPTA-AM largely prevented these effects. HTV activated the IRE1α and PERK arms of the ER stress signaling response and induced mitochondrial dysfunction-NLRP3 inflammasome activation in an IP3R-dependent manner. Similarly, disruption of IP3R/Ca2+ in MLE12 and RAW264.7 cells using carbachol lead to inflammatory responses, and stimulated ER stress and mitochondrial dysfunction. Conclusion Increase in IP3R-mediated Ca2+ release is involved in the inflammatory pathophysiology of VILI via ER stress and mitochondrial dysfunction. Antagonizing IP3R/Ca2+ and/or maintaining Ca2+ homeostasis in lung tissue represents a prospective treatment approach for VILI.
Collapse
Affiliation(s)
- Liu Ye
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qi Zeng
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Maoyao Ling
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Riliang Ma
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Haishao Chen
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhao Li
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, China.,Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
24
|
Perspectives on Mitochondria-ER and Mitochondria-Lipid Droplet Contact in Hepatocytes and Hepatic Lipid Metabolism. Cells 2021; 10:cells10092273. [PMID: 34571924 PMCID: PMC8472694 DOI: 10.3390/cells10092273] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that mitochondrion–endoplasmic reticulum (ER) and mitochondrion–lipid droplet (LD) contact sites are critical in regulating lipid metabolism in cells. It is well established that intracellular organelles communicate with each other continuously through membrane contact sites to maintain organelle function and cellular homeostasis. The accumulation of LDs in hepatocytes is an early indicator of non-alcoholic fatty liver disease (NAFLD) and alcohol-related liver disease (ALD), which may indicate a breakdown in proper inter-organelle communication. In this review, we discuss previous findings in mitochondrion–ER and mitochondrion–LD contact, focusing on their roles in lipid metabolism in hepatocytes. We also present evidence of a unique mitochondrion–LD contact structure in hepatocytes under various physiological and pathological conditions and propose a working hypothesis to speculate about the role of these structures in regulating the functions of mitochondria and LDs and their implications in NAFLD and ALD.
Collapse
|
25
|
Bassot A, Prip-Buus C, Alves A, Berdeaux O, Perrier J, Lenoir V, Ji-Cao J, Berger MA, Loizon E, Cabaret S, Panthu B, Rieusset J, Morio B. Loss and gain of function of Grp75 or mitofusin 2 distinctly alter cholesterol metabolism, but all promote triglyceride accumulation in hepatocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159030. [PMID: 34419589 DOI: 10.1016/j.bbalip.2021.159030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 08/03/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022]
Abstract
In the liver, contact sites between the endoplasmic reticulum (ER) and mitochondria (named MAMs) may be crucial hubs for the regulation of lipid metabolism, thus contributing to the exacerbation or prevention of fatty liver. We hypothesized that tether proteins located at MAMs could play a key role in preventing triglyceride accumulation in hepatocytes and nonalcoholic fatty liver disease (NAFLD) occurrence. To test this, we explored the role of two key partners in building MAM integrity and functionality, the glucose-regulated protein 75 (Grp75) and mitofusin 2 (Mfn2), which liver contents are altered in obesity and NAFLD. Grp75 or Mfn2 expression was either silenced using siRNA or overexpressed with adenoviruses in Huh7 cells. Silencing of Grp75 and Mfn2 resulted in decreased ER-mitochondria interactions, mitochondrial network fusion state and mitochondrial oxidative capacity, while overexpression of the two proteins induced mirror impacts on these parameters. Furthermore, Grp75 or Mfn2 silencing decreased cellular cholesterol content and enhanced triglyceride secretion in ApoB100 lipoproteins, while their overexpression led to reverse effects. Cellular phosphatidylcholine/phosphatidylethanolamine ratio was decreased only upon overexpression of the proteins, potentially contributing to altered ApoB100 assembly and secretion. Despite the opposite differences, both silencing and overexpression of Grp75 or Mfn2 induced triglyceride storage, although a fatty acid challenge was required to express the alteration upon protein silencing. Among the mechanisms potentially involved in this phenotype, ER stress was closely associated with altered triglyceride metabolism after Grp75 or Mfn2 overexpression, while blunted mitochondrial FA oxidation capacity may be the main defect causing triglyceride accumulation upon Grp75 or Mfn2 silencing. Further studies are required to decipher the link between modulation of Grp75 or Mfn2 expression, change in MAM integrity and alteration of cholesterol content of the cell. In conclusion, Grp75 or Mfn2 silencing and overexpression in Huh7 cells contribute to altering MAM integrity and cholesterol storage in opposite directions, but all promote triglyceride accumulation through distinct cellular pathways. This study also highlights that besides Mfn2, Grp75 could play a central role in hepatic lipid and cholesterol metabolism in obesity and NAFLD.
Collapse
Affiliation(s)
- Arthur Bassot
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Carina Prip-Buus
- Institut Cochin, Département d'Endocrinologie, Métabolisme et Diabète, INSERM U1016/CNRS UMR8104/Université de Paris, 75014 Paris, France.
| | - Anaïs Alves
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Olivier Berdeaux
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Université Bourgogne Franche-Comté, Agrosup Dijon, F-21000 Dijon, France.
| | - Johan Perrier
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Véronique Lenoir
- Institut Cochin, Département d'Endocrinologie, Métabolisme et Diabète, INSERM U1016/CNRS UMR8104/Université de Paris, 75014 Paris, France.
| | - Jingwei Ji-Cao
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Marie-Agnès Berger
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Emmanuelle Loizon
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Stephanie Cabaret
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRA, Université Bourgogne Franche-Comté, Agrosup Dijon, F-21000 Dijon, France.
| | - Baptiste Panthu
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Jennifer Rieusset
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| | - Béatrice Morio
- CarMeN Laboratory, INSERM U1060, INRAE U1397, Université Lyon 1, 69008 Lyon, France.
| |
Collapse
|
26
|
Ali ES, Girard D, Petrovsky N. Impaired Ca 2+ signaling due to hepatic steatosis mediates hepatic insulin resistance in Alström syndrome mice that is reversed by GLP-1 analog treatment. Am J Physiol Cell Physiol 2021; 321:C187-C198. [PMID: 34106786 DOI: 10.1152/ajpcell.00020.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ca2+ signaling plays a critical role in the regulation of hepatic metabolism by hormones including insulin. Changes in cytoplasmic Ca2+ regulate synthesis and posttranslational modification of key signaling proteins in the insulin pathways. Emerging evidence suggests that hepatocyte intracellular Ca2+ signaling is altered in lipid-loaded liver cells isolated from obese rodent models. The mechanisms of altered Ca2+-insulin and insulin-Ca2+ signaling pathways in obesity remain poorly understood. Here, we show that the kinetics of insulin-initiated intracellular (initial) Ca2+ release from endoplasmic reticulum is significantly impaired in steatotic hepatocytes from obese Alström syndrome mice. Furthermore, exenatide, a glucagon-like peptide-1 (GLP-1) analog, reversed lipid-induced inhibition of intracellular Ca2+ release kinetics in steatotic hepatocytes, without affecting the total content of intracellular Ca2+ released. Exenatide reversed the lipid-induced inhibition of intracellular Ca2+ release, at least partially, via lipid reduction in hepatocytes, which then restored hormone-regulated cytoplasmic Ca2+ signaling and insulin sensitivity. This data provides additional evidence for the important role of Ca2+ signaling pathways in obesity-associated impaired hepatic lipid homeostasis and insulin signaling. It also highlights a potential advantage of GLP-1 analogs when used to treat type 2 diabetes associated with hepatic steatosis.
Collapse
Affiliation(s)
- Eunus S Ali
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | | | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Vaxine Pty Ltd, Adelaide, South Australia, Australia
| |
Collapse
|
27
|
Zhang L, Zhang Y, Jiang Y, Dou X, Li S, Chai H, Qian Q, Wang M. Upregulated SOCC and IP3R calcium channels and subsequent elevated cytoplasmic calcium signaling promote nonalcoholic fatty liver disease by inhibiting autophagy. Mol Cell Biochem 2021; 476:3163-3175. [PMID: 33864571 DOI: 10.1007/s11010-021-04150-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/01/2021] [Indexed: 12/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is related to elevated cytoplasmic calcium signaling in hepatocytes, which may be mediated by store-operated calcium channel (SOCC) and inositol triphosphate receptor (IP3R). However, the regulatory effect of calcium signaling on lipid accumulation and degeneration in hepatocytes and the underlying molecular mechanism remain unknown. Autophagy inhibition promotes lipid accumulation and steatosis in hepatocytes. However, the association between elevated calcium signaling and autophagy inhibition in hepatocytes and its effect on hepatocyte fatty lesions remain unclear. Here, we established a mouse hepatocyte fatty gradient model using oleic acid. SOCC and IP3R channel opening and cytoplasmic calcium levels gradually increased with the hepatocyte pimelosis degree, whereas autophagy gradually decreased. We also established an optimal oleic acid (OOA) hepatocyte model, observing significantly increased SOCC and IP3R channel opening and calcium influx alongside significantly decreased autophagy and aggravated cellular fatty lesion. Calcium channel blockers (CCBs) and calcium channel gene silencing reagents (CCGSRs), respectively, reversed these effects, indicating that elevated cytoplasmic calcium signaling promotes NAFLD occurrence and the development by inhibiting hepatocyte autophagy. In the OOA model, upregulated extracellular regulated protein kinases 1/2 (ERK1/2), which can be regulated by SOCC and IP3R proteins transient receptor potential canonical 1 (TRPC1)/IP3R with elevated cytoplasmic calcium signaling, over-inhibited forkhead/winged helix O (FOXO) signaling and over-activated mammalian target of rapamycin complex 1 (mTORC1) signaling. Over-inhibited FOXO signaling significantly downregulated autophagy-related gene 12, which inhibits autophagosome maturation, while over-activated mTORC1 signaling over-inactivated Unc-51 like autophagy activating kinase 1, which inhibits preautophagosome formation. CCBs and CCGSRs recovered autophagy by significantly downregulating ERK1/2 to block abnormal changes in FOXO and mTORC1 signaling. Our findings indicate that upregulated SOCC and IP3R channels and subsequent elevated cytoplasmic calcium signaling in hepatocyte fatty lesions inhibits hepatocyte autophagy through (TRPC1/IP3R)/ERK/(FOXO/mTORC1) signaling pathways, causes lipid accumulation and degeneration in hepatocytes, and promotes NAFLD occurrence and development.
Collapse
Affiliation(s)
- Lin Zhang
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yifan Zhang
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Yuanqing Jiang
- College of Life Science, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaobing Dou
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Songtao Li
- Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Hui Chai
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Qianyu Qian
- College of Life Science/Institute of Molecular Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Miaojuan Wang
- Department of General Practice, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
28
|
Jin C, Kumar P, Gracia-Sancho J, Dufour JF. Calcium transfer between endoplasmic reticulum and mitochondria in liver diseases. FEBS Lett 2021; 595:1411-1421. [PMID: 33752262 DOI: 10.1002/1873-3468.14078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/21/2021] [Accepted: 02/25/2021] [Indexed: 01/07/2023]
Abstract
Calcium (Ca2+ ) is a second messenger essential for cellular homeostasis. Inside the cell, Ca2+ is compartmentalized and exchanged among organelles in response to both external and internal stimuli. Mitochondria-associated membranes (MAMs) provide a platform for proteins and channels involved in Ca2+ transfer between the endoplasmic reticulum (ER) and mitochondria. Deregulated Ca2+ signaling and proteins regulating ER-mitochondria interactions have been linked to liver diseases and intensively investigated in recent years. In this review, we summarize the role of MAM-resident proteins in Ca2+ transfer and their association with different liver diseases.
Collapse
Affiliation(s)
- Chaonan Jin
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland
| | - Pavitra Kumar
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland
| | - Jordi Gracia-Sancho
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland.,Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, Barcelona, Spain
| | - Jean-François Dufour
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| |
Collapse
|
29
|
Carpio MA, Means RE, Brill AL, Sainz A, Ehrlich BE, Katz SG. BOK controls apoptosis by Ca 2+ transfer through ER-mitochondrial contact sites. Cell Rep 2021; 34:108827. [PMID: 33691099 PMCID: PMC7995216 DOI: 10.1016/j.celrep.2021.108827] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 11/20/2020] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
Calcium transfer from the endoplasmic reticulum (ER) to mitochondria is a critical contributor to apoptosis. B cell lymphoma 2 (BCL-2) ovarian killer (BOK) localizes to the ER and binds the inositol 1,4,5-trisphosophate receptor (IP3R). Here, we show that BOK is necessary for baseline mitochondrial calcium levels and stimulus-induced calcium transfer from the ER to the mitochondria. Murine embryonic fibroblasts deficient for BOK have decreased proximity of the ER to the mitochondria and altered protein composition of mitochondria-associated membranes (MAMs), which form essential calcium microdomains. Rescue of the ER-mitochondrial juxtaposition with drug-inducible interorganelle linkers reveals a kinetic disruption, which when overcome in Bok−/− cells is still insufficient to rescue thapsigargin-induced calcium transfer and apoptosis. Likewise, a BOK mutant unable to interact with IP3R restores ER-mitochondrial proximity, but not ER-mitochondrial calcium transfer, MAM protein composition, or apoptosis. This work identifies the dynamic coordination of ER-mitochondrial contact by BOK as an important control point for apoptosis. Carpio et al. demonstrate that the proapoptotic BCL-2 family member BOK is present in mitochondrial associated membranes (MAMs). The interaction of BOK with the IP3Rs is critical for its regulation of Ca2+ transfer to the mitochondria, ER-mitochondrial contact sites, and apoptosis.
Collapse
Affiliation(s)
- Marcos A Carpio
- Department of Pathology, Yale School of Medicine, New Haven, CT 06525, USA
| | - Robert E Means
- Department of Pathology, Yale School of Medicine, New Haven, CT 06525, USA
| | - Allison L Brill
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06525, USA
| | - Alva Sainz
- Department of Pathology, Yale School of Medicine, New Haven, CT 06525, USA
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06525, USA; Department of Pharmacology, Yale School of Medicine, New Haven, CT 06525, USA
| | - Samuel G Katz
- Department of Pathology, Yale School of Medicine, New Haven, CT 06525, USA.
| |
Collapse
|
30
|
Mitochondrial metabolism and calcium homeostasis in the development of NAFLD leading to hepatocellular carcinoma. Mitochondrion 2021; 58:24-37. [PMID: 33581332 DOI: 10.1016/j.mito.2021.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a metabolic syndrome characterized by excessive accumulation of hepatic lipid droplets. The disease progresses with steatosis as the premise for hepatocytic damage and tissue scarring, often culminating in hepatocellular carcinoma (HCC). Perturbations in mitochondrial metabolism and energetics were found to be associated with, and often instrumental in various stages of this progression. Functional impairment of the mitochondria affects all aspects of cellular functioning and a particularly important one is calcium signalling. Changes in mitochondrial calcium specifically in hepatocytes of a fatty liver, is reflected by alterations in calcium signalling as well as calcium transporter activities. This deranged Ca2+ homeostasis aids in even more uptake of lipids into the mitochondria and a shift in equilibrium, both metabolically as well as in terms of energy production, leading to completely altered cellular states. These alterations have been reviewed as a perspective to understand the disease progression through NAFLD leading to HCC.
Collapse
|
31
|
Lemmer IL, Willemsen N, Hilal N, Bartelt A. A guide to understanding endoplasmic reticulum stress in metabolic disorders. Mol Metab 2021; 47:101169. [PMID: 33484951 PMCID: PMC7887651 DOI: 10.1016/j.molmet.2021.101169] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/08/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The global rise of metabolic disorders, such as obesity, type 2 diabetes, and cardiovascular disease, demands a thorough molecular understanding of the cellular mechanisms that govern health or disease. The endoplasmic reticulum (ER) is a key organelle for cellular function and metabolic adaptation and, therefore disturbed ER function, known as "ER stress," is a key feature of metabolic disorders. SCOPE OF REVIEW As ER stress remains a poorly defined phenomenon, this review provides a general guide to understanding the nature, etiology, and consequences of ER stress in metabolic disorders. We define ER stress by its type of stressor, which is driven by proteotoxicity, lipotoxicity, and/or glucotoxicity. We discuss the implications of ER stress in metabolic disorders by reviewing evidence implicating ER phenotypes and organelle communication, protein quality control, calcium homeostasis, lipid and carbohydrate metabolism, and inflammation as key mechanisms in the development of ER stress and metabolic dysfunction. MAJOR CONCLUSIONS In mammalian biology, ER is a phenotypically and functionally diverse platform for nutrient sensing, which is critical for cell type-specific metabolic control by hepatocytes, adipocytes, muscle cells, and neurons. In these cells, ER stress is a distinct, transient state of functional imbalance, which is usually resolved by the activation of adaptive programs such as the unfolded protein response (UPR), ER-associated protein degradation (ERAD), or autophagy. However, challenges to proteostasis also impact lipid and glucose metabolism and vice versa. In the ER, sensing and adaptive measures are integrated and failure of the ER to adapt leads to aberrant metabolism, organelle dysfunction, insulin resistance, and inflammation. In conclusion, the ER is intricately linked to a wide spectrum of cellular functions and is a critical component in maintaining and restoring metabolic health.
Collapse
Affiliation(s)
- Imke L Lemmer
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany
| | - Nienke Willemsen
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Nazia Hilal
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Pettenkoferstr. 9, Ludwig-Maximilians-University, 80336 Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Technische Universität München, Biedersteiner Str. 29, 80802 München, Germany; Department of Molecular Metabolism, 665 Huntington Avenue, Harvard T.H. Chan School of Public Health, 02115 Boston, MA, USA.
| |
Collapse
|
32
|
Gaspers LD, Thomas AP, Hoek JB, Bartlett PJ. Ethanol Disrupts Hormone-Induced Calcium Signaling in Liver. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab002. [PMID: 33604575 PMCID: PMC7875097 DOI: 10.1093/function/zqab002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023]
Abstract
Receptor-coupled phospholipase C (PLC) is an important target for the actions of ethanol. In the ex vivo perfused rat liver, concentrations of ethanol >100 mM were required to induce a rise in cytosolic calcium (Ca2+) suggesting that these responses may only occur after binge ethanol consumption. Conversely, pharmacologically achievable concentrations of ethanol (≤30 mM) decreased the frequency and magnitude of hormone-stimulated cytosolic and nuclear Ca2+ oscillations and the parallel translocation of protein kinase C-β to the membrane. Ethanol also inhibited gap junction communication resulting in the loss of coordinated and spatially organized intercellular Ca2+ waves in hepatic lobules. Increasing the hormone concentration overcame the effects of ethanol on the frequency of Ca2+ oscillations and amplitude of the individual Ca2+ transients; however, the Ca2+ responses in the intact liver remained disorganized at the intercellular level, suggesting that gap junctions were still inhibited. Pretreating hepatocytes with an alcohol dehydrogenase inhibitor suppressed the effects of ethanol on hormone-induced Ca2+ increases, whereas inhibiting aldehyde dehydrogenase potentiated the inhibitory actions of ethanol, suggesting that acetaldehyde is the underlying mediator. Acute ethanol intoxication inhibited the rate of rise and the magnitude of hormone-stimulated production of inositol 1,4,5-trisphosphate (IP3), but had no effect on the size of Ca2+ spikes induced by photolysis of caged IP3. These findings suggest that ethanol inhibits PLC activity, but does not affect IP3 receptor function. We propose that by suppressing hormone-stimulated PLC activity, ethanol interferes with the dynamic modulation of [IP3] that is required to generate large, amplitude Ca2+ oscillations.
Collapse
Affiliation(s)
- Lawrence D Gaspers
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA,Address correspondence to L.D.G. (e-mail: )
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jan B Hoek
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paula J Bartlett
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
33
|
Src-mediated Tyr353 phosphorylation of IP3R1 promotes its stability and causes apoptosis in palmitic acid-treated hepatocytes. Exp Cell Res 2021; 399:112438. [PMID: 33358861 DOI: 10.1016/j.yexcr.2020.112438] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 11/01/2020] [Accepted: 12/12/2020] [Indexed: 12/14/2022]
Abstract
Palmitic acid (PA)-induced hepatocyte apoptosis is critical for the progression of nonalcoholic fatty liver disease (NAFLD). Inositol 1,4,5-trisphosphate receptor type 1 (IP3R1) is an intracellular Ca2+-release channel and is involved in PA-induced hepatocyte apoptosis. While the expression of IP3R1 is elevated in patients with NAFLD and in hepatocytes treated with PA, it remains unclear how PA promotes the expression of IP3R1. In present study, our results showed that PA induced mitochondrial dysfunction and apoptosis, which is accompanied with the increase of the IP3R1 expression in hepatic cells. The inhibition of IP3R1 expression using siRNA ameliorated the PA-induced mitochondrial dysfunction. Furthermore, PA enhanced the stability of the IP3R1 protein instead of an increase in its mRNA levels. PA also promoted the phosphorylation of IP3R1 at the Tyr353 site and increased the phosphorylation of src in hepatic cells. Moreover, an inhibitor of src kinase (SU6656) significantly reduced the Tyr353 phosphorylation of IP3R1 and decreased its stability. In addition, SU6656 improved mitochondrial function and reduced apoptosis in hepatocytes. Conclusion: PA promotes the Tyr353 phosphorylation of IP3R1 by activating the src pathway and increasing the protein stability of IP3R1, which consequently results in mitochondrial Ca2+ overload and mitochondrial dysfunction in hepatic cells. Our results also suggested that inhibition of the src/IP3R1 pathway, such as by SU6656, may be a novel potential therapeutic approach for the treatment of NAFLD.
Collapse
|
34
|
Parys JB, Bultynck G, Vervliet T. IP 3 Receptor Biology and Endoplasmic Reticulum Calcium Dynamics in Cancer. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:215-237. [PMID: 34050869 DOI: 10.1007/978-3-030-67696-4_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Intracellular Ca2+ signaling regulates a plethora of cellular functions. A central role in these processes is reserved for the inositol 1,4,5-trisphosphate receptor (IP3R), a ubiquitously expressed Ca2+-release channel, mainly located in the endoplasmic reticulum (ER). Three IP3R isoforms (IP3R1, IP3R2 and IP3R3) exist, encoded respectively by ITPR1, ITPR2 and ITPR3. The proteins encoded by these genes are each about 2700 amino acids long and assemble into large tetrameric channels, which form the target of many regulatory proteins, including several tumor suppressors and oncogenes. Due to the important role of the IP3Rs in cell function, their dysregulation is linked to multiple pathologies. In this review, we highlight the complex role of the IP3R in cancer, as it participates in most of the so-called "hallmarks of cancer". In particular, the IP3R directly controls cell death and cell survival decisions via regulation of autophagy and apoptosis. Moreover, the IP3R impacts cellular proliferation, migration and invasion. Typical examples of the role of the IP3Rs in these various processes are discussed. The relative levels of the IP3R isoforms expressed and their subcellular localization, e.g. at the ER-mitochondrial interface, is hereby important. Finally, evidence is provided about how the knowledge of the regulation of the IP3R by tumor suppressors and oncogenes can be exploited to develop novel therapeutic approaches to fight cancer.
Collapse
Affiliation(s)
- Jan B Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Cancer Institute, KU Leuven, Leuven, Belgium.
| | - Geert Bultynck
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Tim Vervliet
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine & Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Morio B, Panthu B, Bassot A, Rieusset J. Role of mitochondria in liver metabolic health and diseases. Cell Calcium 2020; 94:102336. [PMID: 33387847 DOI: 10.1016/j.ceca.2020.102336] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
The liver is a major organ that coordinates the metabolic flexibility of the whole body, which is characterized by the ability to adapt dynamically in response to fluctuations in energy needs and supplies. In this context, hepatocyte mitochondria are key partners in fine-tuning metabolic flexibility. Here we review the metabolic and signalling pathways carried by mitochondria in the liver, the major pathways that regulate mitochondrial function and how they function in health and metabolic disorders associated to obesity, i.e. insulin resistance, non-alcoholic steatosis and steatohepatitis and hepatocellular carcinoma. Finally, strategies targeting mitochondria to counteract liver disorders are discussed.
Collapse
Affiliation(s)
- Béatrice Morio
- CarMeN Laboratory, INSERM U1060, INRA U1397, Lyon, France
| | | | - Arthur Bassot
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, T6G2H7, Canada
| | | |
Collapse
|
36
|
Townsend LK, Brunetta HS, Mori MAS. Mitochondria-associated ER membranes in glucose homeostasis and insulin resistance. Am J Physiol Endocrinol Metab 2020; 319:E1053-E1060. [PMID: 32985254 DOI: 10.1152/ajpendo.00271.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Obesity and insulin resistance (IR) are associated with endoplasmic reticulum (ER) stress and mitochondrial dysfunction in several tissues. Although for many years mitochondrial and ER function were studied separately, these organelles also connect to produce interdependent functions. Communication occurs at mitochondria-associated ER membranes (MAMs) and regulates lipid and calcium homeostasis, apoptosis, and the exchange of adenine nucleotides, among other things. Recent evidence suggests that MAMs contribute to organelle, cellular, and systemic metabolism. In obesity and IR models, metabolic tissues such as the liver, skeletal muscle, pancreas, and adipose tissue present alterations in MAM structure or function. The purpose of this mini review is to highlight the MAM disruptions that occur in each tissue during obesity and IR and its relationship with glucose homeostasis and IR. We also discuss the current controversy that surrounds MAMs' role in the development of IR.
Collapse
Affiliation(s)
- Logan K Townsend
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Henver S Brunetta
- Department of Physiological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Department of Biochemistry and Tissue Biology, University of Campinas, Campinas, Brazil
| | - Marcelo A S Mori
- Department of Biochemistry and Tissue Biology, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster, University of Campinas, Campinas, Brazil
| |
Collapse
|
37
|
Lemos FO, Guerra MT, Leite MDF. Inositol 1,4,5 trisphosphate receptors in secretory epithelial cells of the gastrointestinal tract. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Lima Filho ACM, França A, Florentino RM, Dos Santos ML, de Oliveira Lemos F, Missiaggia DG, Fonseca RC, Gustavo Oliveira A, Ananthanarayanan M, Guerra MT, de Castro Fonseca M, Vidigal PVT, Lima CX, Nathanson MH, Fatima Leite M. Inositol 1,4,5-trisphosphate receptor type 3 plays a protective role in hepatocytes during hepatic ischemia-reperfusion injury. Cell Calcium 2020; 91:102264. [PMID: 32957029 DOI: 10.1016/j.ceca.2020.102264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 01/02/2023]
Abstract
Hepatic ischemia-reperfusion injury is seen in a variety of clinical conditions, including hepatic thrombosis, systemic hypotension, and liver transplantation. Calcium (Ca2+) signaling mediates several pathophysiological processes in the liver, but it is not known whether and how intracellular Ca2+ channels are involved in the hepatocellular events secondary to ischemia-reperfusion. Using an animal model of hepatic ischemia-reperfusion injury, we observed a progressive increase in expression of the type 3 isoform of the inositol trisphosphate receptor (ITPR3), an intracellular Ca2+ channel that is not normally expressed in healthy hepatocytes. ITPR3 expression was upregulated, at least in part, by a combination of demethylation of the ITPR3 promoter region and the increased transcriptional activity of the nuclear factor of activated T-cells (NFAT). Additionally, expression of pro-inflammatory interleukins and necrotic surface area were less pronounced in livers of control animals compared to liver-specific ITPR3 KO mice subjected to hepatic damage. Corroborating these findings, ITPR3 expression and activation of NFAT were observed in hepatocytes of liver biopsies from patients who underwent liver ischemia caused by thrombosis after organ transplant. Together, these results are consistent with the idea that ITPR3 expression in hepatocytes plays a protective role during hepatic injury induced by ischemia-reperfusion.
Collapse
Affiliation(s)
| | - Andressa França
- Department of Molecular Medicine, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | - Rodrigo M Florentino
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | | | | | | | | | - André Gustavo Oliveira
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | | | - Mateus T Guerra
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, CT, United States.
| | - Matheus de Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials, SP, Brazil.
| | | | - Cristiano Xavier Lima
- Department of Surgery, Medicine School of Federal University of Minas Gerais (UFMG), MG, United States.
| | - Michael H Nathanson
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, CT, United States.
| | - M Fatima Leite
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| |
Collapse
|
39
|
Lemos FDO, França A, Lima Filho ACM, Florentino RM, Santos ML, Missiaggia DG, Rodrigues GOL, Dias FF, Souza Passos IB, Teixeira MM, Andrade AMDF, Lima CX, Vidigal PVT, Costa VV, Fonseca MC, Nathanson MH, Leite MF. Molecular Mechanism for Protection Against Liver Failure in Human Yellow Fever Infection. Hepatol Commun 2020; 4:657-669. [PMID: 32363317 PMCID: PMC7193135 DOI: 10.1002/hep4.1504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Yellow fever (YF) is a viral hemorrhagic fever that typically involves the liver. Brazil recently experienced its largest recorded YF outbreak, and the disease was fatal in more than a third of affected individuals, mostly because of acute liver failure. Affected individuals are generally treated only supportively, but during the recent Brazilian outbreak, selected patients were treated with liver transplant. We took advantage of this clinical experience to better characterize the clinical and pathological features of YF-induced liver failure and to examine the mechanism of hepatocellular injury in YF, to identify targets that would be amenable to therapeutic intervention in preventing progression to liver failure and death. Patients with YF liver failure rapidly developed massive transaminase elevations, with jaundice, coagulopathy, thrombocytopenia, and usually hepatic encephalopathy, along with pathological findings that included microvesicular steatosis and lytic necrosis. Hepatocytes began to express the type 3 isoform of the inositol trisphosphate receptor (ITPR3), an intracellular calcium (Ca2+) channel that is not normally expressed in hepatocytes. Experiments in an animal model, isolated hepatocytes, and liver-derived cell lines showed that this new expression of ITPR3 was associated with increased nuclear Ca2+ signaling and hepatocyte proliferation, and reduced steatosis and cell death induced by the YF virus. Conclusion: Yellow fever often induces liver failure characterized by massive hepatocellular damage plus steatosis. New expression of ITPR3 also occurs in YF-infected hepatocytes, which may represent an endogenous protective mechanism that could suggest approaches to treat affected individuals before they progress to liver failure, thereby decreasing the mortality of this disease in a way that does not rely on the costly and limited resource of liver transplantation.
Collapse
Affiliation(s)
| | - Andressa França
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Rodrigo M. Florentino
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Marcone Loiola Santos
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Dabny G. Missiaggia
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Felipe Ferraz Dias
- Center of MicroscopyUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Mauro M. Teixeira
- Department of Biochemistry and ImmunologyUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Cristiano Xavier Lima
- Hepatic Transplant ServiceHospital Felício RochoBelo HorizonteBrazil
- SurgeryUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | | | - Matheus Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio)Brazilian Center for Research in Energy and MaterialsRua Giuseppe Máximo ScolfaroCampinasBrazil
| | - Michael H. Nathanson
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCT
| | - M. Fatima Leite
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| |
Collapse
|
40
|
Perry RJ, Zhang D, Guerra MT, Brill AL, Goedeke L, Nasiri AR, Rabin-Court A, Wang Y, Peng L, Dufour S, Zhang Y, Zhang XM, Butrico GM, Toussaint K, Nozaki Y, Cline GW, Petersen KF, Nathanson MH, Ehrlich BE, Shulman GI. Glucagon stimulates gluconeogenesis by INSP3R1-mediated hepatic lipolysis. Nature 2020; 579:279-283. [PMID: 32132708 PMCID: PMC7101062 DOI: 10.1038/s41586-020-2074-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 01/15/2020] [Indexed: 11/09/2022]
Abstract
While it is well-established that alterations in the portal vein insulin/glucagon ratio play a major role in causing dysregulated hepatic glucose metabolism in type 2 diabetes (T2D)1–3, the mechanisms by which glucagon alters hepatic glucose production and mitochondrial oxidation remain poorly understood. Here we show that glucagon stimulates hepatic gluconeogenesis by increasing hepatic adipose triglyceride lipase activity, intrahepatic lipolysis, hepatic acetyl-CoA content, and pyruvate carboxylase flux, while also increasing mitochondrial fat oxidation, mediated by stimulation of the inositol triphosphate receptor-1 (InsP3R-I). Chronic physiological increases in plasma glucagon concentrations increased mitochondrial hepatic fat oxidation and reversed diet-induced hepatic steatosis and insulin resistance in rats and mice; however, the effect of chronic glucagon treatment to reverse hepatic steatosis and glucose intolerance was abrogated in InsP3R-I knockout mice. These results provide new insights into glucagon biology and suggest that InsP3R-I may be a novel therapeutic target to reverse nonalcoholic fatty liver disease and T2D.
Collapse
Affiliation(s)
- Rachel J Perry
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Dongyan Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Mateus T Guerra
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Allison L Brill
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Leigh Goedeke
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ali R Nasiri
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Aviva Rabin-Court
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Yongliang Wang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Liang Peng
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sylvie Dufour
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ye Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Xian-Man Zhang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Gina M Butrico
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Keshia Toussaint
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Yuichi Nozaki
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Gary W Cline
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Michael H Nathanson
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA.,Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA. .,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
41
|
Alogaili F, Chinnarasu S, Jaeschke A, Kranias EG, Hui DY. Hepatic HAX-1 inactivation prevents metabolic diseases by enhancing mitochondrial activity and bile salt export. J Biol Chem 2020; 295:4631-4646. [PMID: 32079675 DOI: 10.1074/jbc.ra119.012361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/04/2020] [Indexed: 12/26/2022] Open
Abstract
Increasing hepatic mitochondrial activity through pyruvate dehydrogenase and elevating enterohepatic bile acid recirculation are promising new approaches for metabolic disease therapy, but neither approach alone can completely ameliorate disease phenotype in high-fat diet-fed mice. This study showed that diet-induced hepatosteatosis, hyperlipidemia, and insulin resistance can be completely prevented in mice with liver-specific HCLS1-associated protein X-1 (HAX-1) inactivation. Mechanistically, we showed that HAX-1 interacts with inositol 1,4,5-trisphosphate receptor-1 (InsP3R1) in the liver, and its absence reduces InsP3R1 levels, thereby improving endoplasmic reticulum-mitochondria calcium homeostasis to prevent excess calcium overload and mitochondrial dysfunction. As a result, HAX-1 ablation activates pyruvate dehydrogenase and increases mitochondria utilization of glucose and fatty acids to prevent hepatosteatosis, hyperlipidemia, and insulin resistance. In contrast to the reduction of InsP3R1 levels, hepatic HAX-1 deficiency increases bile salt exporter protein levels, thereby promoting enterohepatic bile acid recirculation, leading to activation of bile acid-responsive genes in the intestinal ileum to augment insulin sensitivity and of cholesterol transport genes in the liver to suppress hyperlipidemia. The dual mechanisms of increased mitochondrial respiration and enterohepatic bile acid recirculation due to improvement of endoplasmic reticulum-mitochondria calcium homeostasis with hepatic HAX-1 inactivation suggest that this may be a potential therapeutic target for metabolic disease intervention.
Collapse
Affiliation(s)
- Fawzi Alogaili
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Sivaprakasam Chinnarasu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| |
Collapse
|
42
|
Ueasilamongkol P, Khamphaya T, Guerra MT, Rodrigues M, Gomes DA, Kong Y, Wei W, Jain D, Trampert DC, Ananthanarayanan M, Banales JM, Roberts LR, Farshidfar F, Nathanson MH, Weerachayaphorn J. Type 3 Inositol 1,4,5-Trisphosphate Receptor Is Increased and Enhances Malignant Properties in Cholangiocarcinoma. Hepatology 2020; 71:583-599. [PMID: 31251815 PMCID: PMC6934938 DOI: 10.1002/hep.30839] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
Cholangiocarcinoma (CCA) is the second most common malignancy arising in the liver. It carries a poor prognosis, in part because its pathogenesis is not well understood. The type 3 inositol 1,4,5-trisphosphate receptor (ITPR3) is the principal intracellular calcium ion (Ca2+ ) release channel in cholangiocytes, and its increased expression has been related to the pathogenesis of malignancies in other types of tissues, so we investigated its role in CCA. ITPR3 expression was increased in both hilar and intrahepatic CCA samples as well as in CCA cell lines. Deletion of ITPR3 from CCA cells impaired proliferation and cell migration. A bioinformatic analysis suggested that overexpression of ITPR3 in CCA would have a mitochondrial phenotype, so this was also examined. ITPR3 normally is concentrated in a subapical region of endoplasmic reticulum (ER) in cholangiocytes, but both immunogold electron microscopy and super-resolution microscopy showed that ITPR3 in CCA cells was also in regions of ER in close association with mitochondria. Deletion of ITPR3 from these cells impaired mitochondrial Ca2+ signaling and led to cell death. Conclusion: ITPR3 expression in cholangiocytes becomes enhanced in CCA. This contributes to malignant features, including cell proliferation and migration and enhanced mitochondrial Ca2+ signaling.
Collapse
Affiliation(s)
| | - Tanaporn Khamphaya
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Mateus T. Guerra
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michele Rodrigues
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dawidson A. Gomes
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yong Kong
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Wei Wei
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Dhanpat Jain
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - David C. Trampert
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
| | - Lewis R. Roberts
- Divisions of Gastroenterology and Hepatology and Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Farshad Farshidfar
- Department of Oncology, Cumming School of Medicine, University of Calgary, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Michael H. Nathanson
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jittima Weerachayaphorn
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
43
|
Lemos FDO, Florentino RM, Lima Filho ACM, Santos MLD, Leite MF. Inositol 1,4,5-trisphosphate receptor in the liver: Expression and function. World J Gastroenterol 2019; 25:6483-6494. [PMID: 31802829 PMCID: PMC6886013 DOI: 10.3748/wjg.v25.i44.6483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is a complex organ that performs several functions to maintain homeostasis. These functions are modulated by calcium, a second messenger that regulates several intracellular events. In hepatocytes and cholangiocytes, which are the epithelial cell types in the liver, inositol 1,4,5-trisphosphate (InsP3) receptors (ITPR) are the only intracellular calcium release channels. Three isoforms of the ITPR have been described, named type 1, type 2 and type 3. These ITPR isoforms are differentially expressed in liver cells where they regulate distinct physiological functions. Changes in the expression level of these receptors correlate with several liver diseases and hepatic dysfunctions. In this review, we highlight how the expression level, modulation, and localization of ITPR isoforms in hepatocytes and cholangiocytes play a role in hepatic homeostasis and liver pathology.
Collapse
Affiliation(s)
- Fernanda de Oliveira Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Rodrigo M Florentino
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Antônio Carlos Melo Lima Filho
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Marcone Loiola dos Santos
- Department of Cell Biology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - M Fatima Leite
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| |
Collapse
|
44
|
Type 3 inositol 1,4,5-trisphosphate receptor: A calcium channel for all seasons. Cell Calcium 2019; 85:102132. [PMID: 31790953 DOI: 10.1016/j.ceca.2019.102132] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022]
Abstract
Inositol 1,4,5 trisphosphate receptors (ITPRs) are a family of endoplasmic reticulum Ca2+ channels essential for the control of intracellular Ca2+ levels in virtually every mammalian cell type. The three isoforms (ITPR1, ITPR2 and ITPR3) are highly homologous in amino acid sequence, but they differ considerably in terms of biophysical properties, subcellular localization, and tissue distribution. Such differences underscore the variety of cellular responses triggered by each isoform and suggest that the expression/activity of specific isoforms might be linked to particular pathophysiological states. Indeed, recent findings demonstrate that changes in expression of ITPR isoforms are associated with a number of human diseases ranging from fatty liver disease to cancer. ITPR3 is emerging as the isoform that is particularly important in the pathogenesis of various human diseases. Here we review the physiological and pathophysiological roles of ITPR3 in various tissues and the mechanisms by which the expression of this isoform is modulated in health and disease.
Collapse
|
45
|
Regulation of Mitochondria-Associated Membranes (MAMs) by NO/sGC/PKG Participates in the Control of Hepatic Insulin Response. Cells 2019; 8:cells8111319. [PMID: 31731523 PMCID: PMC6912364 DOI: 10.3390/cells8111319] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/07/2019] [Accepted: 10/22/2019] [Indexed: 12/13/2022] Open
Abstract
Under physiological conditions, nitric oxide (NO) produced by the endothelial NO synthase (eNOS) upregulates hepatic insulin sensitivity. Recently, contact sites between the endoplasmic reticulum and mitochondria named mitochondria-associated membranes (MAMs) emerged as a crucial hub for insulin signaling in the liver. As mitochondria are targets of NO, we explored whether NO regulates hepatic insulin sensitivity by targeting MAMs. In Huh7 cells, primary rat hepatocytes and mouse livers, enhancing NO concentration increased MAMs, whereas inhibiting eNOS decreased them. In vitro, those effects were prevented by inhibiting protein kinase G (PKG) and mimicked by activating soluble guanylate cyclase (sGC) and PKG. In agreement with the regulation of MAMs, increasing NO concentration improved insulin signaling, both in vitro and in vivo, while eNOS inhibition disrupted this response. Finally, inhibition of insulin signaling by wortmannin did not affect the impact of NO on MAMs, while experimental MAM disruption, using either targeted silencing of cyclophilin D or the overexpression of the organelle spacer fetal and adult testis-expressed 1 (FATE-1), significantly blunted the effects of NO on both MAMs and insulin response. Therefore, under physiological conditions, NO participates to the regulation of MAM integrity through the sGC/PKG pathway and concomitantly improves hepatic insulin sensitivity. Altogether, our data suggest that the induction of MAMs participate in the impact of NO on hepatocyte insulin response.
Collapse
|
46
|
Guerra MT, Florentino RM, Franca A, Filho ACL, dos Santos ML, Fonseca RC, Lemos FO, Fonseca MC, Kruglov E, Mennone A, Njei B, Gibson J, Guan F, Cheng YC, Ananthanarayanam M, Gu J, Jiang J, Zhao H, Lima CX, Vidigal PT, Oliveira AG, Nathanson MH, Leite MF. Expression of the type 3 InsP 3 receptor is a final common event in the development of hepatocellular carcinoma. Gut 2019; 68:1676-1687. [PMID: 31315892 PMCID: PMC7087395 DOI: 10.1136/gutjnl-2018-317811] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/25/2019] [Accepted: 06/30/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & OBJECTIVES Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. Several types of chronic liver disease predispose to HCC, and several different signalling pathways have been implicated in its pathogenesis, but no common molecular event has been identified. Ca2+ signalling regulates the proliferation of both normal hepatocytes and liver cancer cells, so we investigated the role of intracellular Ca2+ release channels in HCC. DESIGN Expression analyses of the type 3 isoform of the inositol 1, 4, 5-trisphosphate receptor (ITPR3) in human liver samples, liver cancer cells and mouse liver were combined with an evaluation of DNA methylation profiles of ITPR3 promoter in HCC and characterisation of the effects of ITPR3 expression on cellular proliferation and apoptosis. The effects of de novo ITPR3 expression on hepatocyte calcium signalling and liver growth were evaluated in mice. RESULTS ITPR3 was absent or expressed in low amounts in hepatocytes from normal liver, but was expressed in HCC specimens from three independent patient cohorts, regardless of the underlying cause of chronic liver disease, and its increased expression level was associated with poorer survival. The ITPR3 gene was heavily methylated in control liver specimens but was demethylated at multiple sites in specimens of patient with HCC. Administration of a demethylating agent in a mouse model resulted in ITPR3 expression in discrete areas of the liver, and Ca2+ signalling was enhanced in these regions. In addition, cell proliferation and liver regeneration were enhanced in the mouse model, and deletion of ITPR3 from human HCC cells enhanced apoptosis. CONCLUSIONS These results provide evidence that de novo expression of ITPR3 typically occurs in HCC and may play a role in its pathogenesis.
Collapse
MESH Headings
- Adult
- Animals
- Apoptosis/physiology
- Calcium Signaling/physiology
- Carcinogenesis/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Proliferation/physiology
- Cells, Cultured
- DNA Methylation
- Female
- Gene Expression Regulation, Neoplastic/physiology
- Hepatocytes/metabolism
- Humans
- Inositol 1,4,5-Trisphosphate Receptors/deficiency
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Liver/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Regeneration/physiology
- Male
- Mice, Knockout
- Middle Aged
- Survival Analysis
Collapse
Affiliation(s)
- Mateus T Guerra
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rodrigo M Florentino
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andressa Franca
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antonio C Lima Filho
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcone L dos Santos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Roberta C Fonseca
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda O Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus C Fonseca
- Centro Nacional de Pesquisa em Energia e Materiais, Campinas, Brazil
| | - Emma Kruglov
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert Mennone
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Basile Njei
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joanna Gibson
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Fulan Guan
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jianlei Gu
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, China
| | - Jianping Jiang
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Cristiano X Lima
- Department of Surgery, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula T Vidigal
- Department of Pathological Anatomy and Forensic Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andre G Oliveira
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michael H Nathanson
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maria Fatima Leite
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
47
|
Ali ES, Rychkov GY, Barritt GJ. Deranged hepatocyte intracellular Ca 2+ homeostasis and the progression of non-alcoholic fatty liver disease to hepatocellular carcinoma. Cell Calcium 2019; 82:102057. [PMID: 31401389 DOI: 10.1016/j.ceca.2019.102057] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths in men, and the sixth in women. Non-alcoholic fatty liver disease (NAFLD) is now one of the major risk factors for HCC. NAFLD, which involves the accumulation of excess lipid in cytoplasmic lipid droplets in hepatocytes, can progress to non-alcoholic steatosis, fibrosis, and HCC. Changes in intracellular Ca2+ constitute important signaling pathways for the regulation of lipid and carbohydrate metabolism in normal hepatocytes. Recent studies of steatotic hepatocytes have identified lipid-induced changes in intracellular Ca2+, and have provided evidence that altered Ca2+ signaling exacerbates lipid accumulation and may promote HCC. The aims of this review are to summarise current knowledge of the lipid-induced changes in hepatocyte Ca2+ homeostasis, to comment on the mechanisms involved, and discuss the pathways leading from altered Ca2+ homeostasis to enhanced lipid accumulation and the potential promotion of HCC. In steatotic hepatocytes, lipid inhibits store-operated Ca2+ entry and SERCA2b, and activates Ca2+ efflux from the endoplasmic reticulum (ER) and its transfer to mitochondria. These changes are associated with changes in Ca2+ concentrations in the ER (decreased), cytoplasmic space (increased) and mitochondria (likely increased). They lead to: inhibition of lipolysis, lipid autophagy, lipid oxidation, and lipid secretion; activation of lipogenesis; increased lipid; ER stress, generation of reactive oxygen species (ROS), activation of Ca2+/calmodulin-dependent kinases and activation of transcription factor Nrf2. These all can potentially mediate the transition of NAFLD to HCC. It is concluded that lipid-induced changes in hepatocyte Ca2+ homeostasis are important in the initiation and progression of HCC. Further research is desirable to better understand the cause and effect relationships, the time courses and mechanisms involved, and the potential of Ca2+ transporters, channels, and binding proteins as targets for pharmacological intervention.
Collapse
Affiliation(s)
- Eunus S Ali
- Department of Medical Biochemistry, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, 5001, Australia
| | - Grigori Y Rychkov
- School of Medicine, The University of Adelaide, and South Australian Health and Medical Research Institute, Adelaide, South Australia, 5005, Australia
| | - Greg J Barritt
- Department of Medical Biochemistry, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, 5001, Australia.
| |
Collapse
|
48
|
Ali ES, Petrovsky N. Calcium Signaling As a Therapeutic Target for Liver Steatosis. Trends Endocrinol Metab 2019; 30:270-281. [PMID: 30850262 DOI: 10.1016/j.tem.2019.02.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022]
Abstract
Hepatic steatosis, the first step in nonalcoholic fatty liver disease (NAFLD), can arise from various pathophysiological conditions. While lipid metabolism in the liver is normally balanced such that there is no excessive lipid accumulation, when this homeostasis is disrupted lipid droplets (LDs) accumulate in hepatocytes resulting in cellular toxicity. The mechanisms underlying this accumulation and the subsequent hepatocellular damage are multifactorial and poorly understood, with the result that there are no currently approved treatments for NAFLD. Impaired calcium signaling has recently been identified as a cause of increased endoplasmic reticulum (ER) stress contributing to hepatic lipid accumulation. This review highlights new findings on the role of impaired Ca2+ signaling in the development of steatosis and discusses potential new approaches to NAFLD treatment based on these new insights.
Collapse
Affiliation(s)
- Eunüs S Ali
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Adelaide, SA, Australia.
| |
Collapse
|
49
|
Thoudam T, Ha CM, Leem J, Chanda D, Park JS, Kim HJ, Jeon JH, Choi YK, Liangpunsakul S, Huh YH, Kwon TH, Park KG, Harris RA, Park KS, Rhee HW, Lee IK. PDK4 Augments ER-Mitochondria Contact to Dampen Skeletal Muscle Insulin Signaling During Obesity. Diabetes 2019; 68:571-586. [PMID: 30523025 PMCID: PMC6385748 DOI: 10.2337/db18-0363] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 11/20/2018] [Indexed: 12/17/2022]
Abstract
Mitochondria-associated endoplasmic reticulum membrane (MAM) is a structural link between mitochondria and endoplasmic reticulum (ER). MAM regulates Ca2+ transport from the ER to mitochondria via an IP3R1-GRP75-VDAC1 complex-dependent mechanism. Excessive MAM formation may cause mitochondrial Ca2+ overload and mitochondrial dysfunction. However, the exact implication of MAM formation in metabolic syndromes remains debatable. Here, we demonstrate that PDK4 interacts with and stabilizes the IP3R1-GRP75-VDAC1 complex at the MAM interface. Obesity-induced increase in PDK4 activity augments MAM formation and suppresses insulin signaling. Conversely, PDK4 inhibition dampens MAM formation and improves insulin signaling by preventing MAM-induced mitochondrial Ca2+ accumulation, mitochondrial dysfunction, and ER stress. Furthermore, Pdk4-/- mice exhibit reduced MAM formation and are protected against diet-induced skeletal muscle insulin resistance. Finally, forced formation and stabilization of MAMs with synthetic ER-mitochondria linker prevented the beneficial effects of PDK4 deficiency on insulin signaling. Overall, our findings demonstrate a critical mediatory role of PDK4 in the development of skeletal muscle insulin resistance via enhancement of MAM formation.
Collapse
Affiliation(s)
- Themis Thoudam
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Chae-Myeong Ha
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Dipanjan Chanda
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jong-Seok Park
- Department of Chemistry, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Hyo-Jeong Kim
- Electron Microscopy Research Center, Korea Basic Science Institute, Ochang, Chungbuk, Republic of Korea
| | - Jae-Han Jeon
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yeon-Kyung Choi
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush VA Medical Center, Indianapolis, IN
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Ochang, Chungbuk, Republic of Korea
| | - Tae-Hwan Kwon
- Department of Biomedical Science, The Graduate School, Kyungpook National University, Daegu, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, Kyungpook National University, Daegu, Republic of Korea
| | - Keun-Gyu Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Robert A Harris
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Kyu-Sang Park
- Department of Physiology, Institute of Lifestyle Medicine, Yonsei University Wonju College of Medicine, Gangwon-Do, Republic of Korea
| | - Hyun-Woo Rhee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - In-Kyu Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
50
|
Rodrigues MA, Gomes DA, Nathanson MH. Calcium Signaling in Cholangiocytes: Methods, Mechanisms, and Effects. Int J Mol Sci 2018; 19:ijms19123913. [PMID: 30563259 PMCID: PMC6321159 DOI: 10.3390/ijms19123913] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
Calcium (Ca2+) is a versatile second messenger that regulates a number of cellular processes in virtually every type of cell. The inositol 1,4,5-trisphosphate receptor (ITPR) is the only intracellular Ca2+ release channel in cholangiocytes, and is therefore responsible for Ca2+-mediated processes in these cells. This review will discuss the machinery responsible for Ca2+ signals in these cells, as well as experimental models used to investigate cholangiocyte Ca2+ signaling. We will also discuss the role of Ca2+ in the normal and abnormal regulation of secretion and apoptosis in cholangiocytes, two of the best characterized processes mediated by Ca2+ in this cell type.
Collapse
Affiliation(s)
- Michele Angela Rodrigues
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
| | - Dawidson Assis Gomes
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
- Department of Biochemistry and Immunology, Federal University of Minas Gerais. Av. Antônio Carlos, 6627, Belo Horizonte-MG 31270-901, Brazil.
| | - Michael Harris Nathanson
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
| |
Collapse
|