51
|
Florance I, Ramasubbu S. Current Understanding on the Role of Lipids in Macrophages and Associated Diseases. Int J Mol Sci 2022; 24:ijms24010589. [PMID: 36614031 PMCID: PMC9820199 DOI: 10.3390/ijms24010589] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/31/2022] Open
Abstract
Lipid metabolism is the major intracellular mechanism driving a variety of cellular functions such as energy storage, hormone regulation and cell division. Lipids, being a primary component of the cell membrane, play a pivotal role in the survival of macrophages. Lipids are crucial for a variety of macrophage functions including phagocytosis, energy balance and ageing. However, functions of lipids in macrophages vary based on the site the macrophages are residing at. Lipid-loaded macrophages have recently been emerging as a hallmark for several diseases. This review discusses the significance of lipids in adipose tissue macrophages, tumor-associated macrophages, microglia and peritoneal macrophages. Accumulation of macrophages with impaired lipid metabolism is often characteristically observed in several metabolic disorders. Stress signals differentially regulate lipid metabolism. While conditions such as hypoxia result in accumulation of lipids in macrophages, stress signals such as nutrient deprivation initiate lipolysis and clearance of lipids. Understanding the biology of lipid accumulation in macrophages requires the development of potentially active modulators of lipid metabolism.
Collapse
|
52
|
Schultz CW, Nevler A. Pyrvinium Pamoate: Past, Present, and Future as an Anti-Cancer Drug. Biomedicines 2022; 10:3249. [PMID: 36552005 PMCID: PMC9775650 DOI: 10.3390/biomedicines10123249] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022] Open
Abstract
Pyrvinium, a lipophilic cation belonging to the cyanine dye family, has been used in the clinic as a safe and effective anthelminthic for over 70 years. Its structure, similar to some polyaminopyrimidines and mitochondrial-targeting peptoids, has been linked with mitochondrial localization and targeting. Over the past two decades, increasing evidence has emerged showing pyrvinium to be a strong anti-cancer molecule in various human cancers in vitro and in vivo. This efficacy against cancers has been attributed to diverse mechanisms of action, with the weight of evidence supporting the inhibition of mitochondrial function, the WNT pathway, and cancer stem cell renewal. Despite the overwhelming evidence demonstrating the efficacy of pyrvinium for the treatment of human cancers, pyrvinium has not yet been repurposed for the treatment of cancers. This review provides an in-depth analysis of the history of pyrvinium as a therapeutic, the rationale and data supporting its use as an anticancer agent, and the challenges associated with repurposing pyrvinium as an anti-cancer agent.
Collapse
Affiliation(s)
- Christopher W. Schultz
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Avinoam Nevler
- Jefferson Pancreas, Biliary, and Related Cancer Center, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
53
|
Kapelanski-Lamoureux A, Chen Z, Gao ZH, Deng R, Lazaris A, Lebeaupin C, Giles L, Malhotra J, Yong J, Zou C, de Jong YP, Metrakos P, Herzog RW, Kaufman RJ. Ectopic clotting factor VIII expression and misfolding in hepatocytes as a cause for hepatocellular carcinoma. Mol Ther 2022; 30:3542-3551. [PMID: 36242517 PMCID: PMC9734080 DOI: 10.1016/j.ymthe.2022.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022] Open
Abstract
Hemophilia A gene therapy targets hepatocytes to express B domain deleted (BDD) clotting factor VIII (FVIII) to permit viral encapsidation. Since BDD is prone to misfolding in the endoplasmic reticulum (ER) and ER protein misfolding in hepatocytes followed by high-fat diet (HFD) can cause hepatocellular carcinoma (HCC), we studied how FVIII misfolding impacts HCC development using hepatocyte DNA delivery to express three proteins from the same parental vector: (1) well-folded cytosolic dihydrofolate reductase (DHFR); (2) BDD-FVIII, which is prone to misfolding in the ER; and (3) N6-FVIII, which folds more efficiently than BDD-FVIII. One week after DNA delivery, when FVIII expression was undetectable, mice were fed HFD for 65 weeks. Remarkably, all mice that received BDD-FVIII vector developed liver tumors, whereas only 58% of mice that received N6 and no mice that received DHFR vector developed liver tumors, suggesting that the degree of protein misfolding in the ER increases predisposition to HCC in the context of an HFD and in the absence of viral transduction. Our findings raise concerns of ectopic BDD-FVIII expression in hepatocytes in the clinic, which poses risks independent of viral vector integration. Limited expression per hepatocyte and/or use of proteins that avoid misfolding may enhance safety.
Collapse
Affiliation(s)
- Audrey Kapelanski-Lamoureux
- Department of Anatomy and Cell Biology, McGill University, Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Zhouji Chen
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Zu-Hua Gao
- Department of Pathology and Oncology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada,Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Ruishu Deng
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Anthoula Lazaris
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Cynthia Lebeaupin
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, SBP Medical Discovery Institute, La Jolla, CA 92037, USA,Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lisa Giles
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jyoti Malhotra
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jing Yong
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Chenhui Zou
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ype P. de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Peter Metrakos
- Department of Surgery, McGill University; Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Randal J. Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, SBP Medical Discovery Institute, La Jolla, CA 92037, USA,Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, MI 48109, USA,Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, USA,Howard Hughes Medical Institute, University of Michigan Medical Center, Ann Arbor, MI 48109, USA,Corresponding author: Randal J. Kaufman, Degenerative Diseases Program, SBP Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
54
|
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]
|
55
|
Magee N, Ahamed F, Eppler N, Jones E, Ghosh P, He L, Zhang Y. Hepatic transcriptome profiling reveals early signatures associated with disease transition from non-alcoholic steatosis to steatohepatitis. LIVER RESEARCH 2022; 6:238-250. [PMID: 36864891 PMCID: PMC9977163 DOI: 10.1016/j.livres.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background and aim Non-alcoholic fatty liver disease (NAFLD) is becoming a leading cause of chronic liver disease worldwide. The molecular events that influence disease progression from non-alcoholic fatty liver (NAFL) to aggressive non-alcoholic steatohepatitis (NASH) remain incompletely understood, leading to lack of mechanism-based targeted treatment options for NASH. This study aims to identify early signatures associated with disease progression from NAFL to NASH in mice and humans. Materials and methods Male C57BL/6J mice were fed a high-fat, -cholesterol, and - fructose (HFCF) diet for up to 9 months. The extent of steatosis, inflammation, and fibrosis was evaluated in liver tissues. Total RNA sequencing (RNA-seq) was conducted to determine liver transcriptomic changes. Results After being fed the HFCF diet, mice sequentially developed steatosis, early steatohepatitis, steatohepatitis with fibrosis, and eventually spontaneous liver tumor. Hepatic RNA-seq revealed that the key signatures during steatosis progression to early steatohepatitis were pathways related to extracellular matrix organization and immune responses such as T cell migration, arginine biosynthesis, C-type lectin receptor signaling, and cytokine-cytokine receptor interaction. Genes regulated by transcription factors forkhead box M1 (FOXM1) and negative elongation factor complex member E (NELFE) were significantly altered during disease progression. This phenomenon was also observed in patients with NASH. Conclusions In summary, we identified early signatures associated with disease progression from NAFL to early NASH in a mouse model that recapitulated key metabolic, histologic, and transcriptomic changes seen in humans. The findings from our study may shed light on the development of novel preventative, diagnostic, and therapeutic strategies for NASH.
Collapse
Affiliation(s)
- Nancy Magee
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Forkan Ahamed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Natalie Eppler
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Elizabeth Jones
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Priyanka Ghosh
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lily He
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Yuxia Zhang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
- Liver Center, University of Kansas, Kansas City, KS, USA
| |
Collapse
|
56
|
Martínez JA, Alonso-Bernáldez M, Martínez-Urbistondo D, Vargas-Nuñez JA, Ramírez de Molina A, Dávalos A, Ramos-Lopez O. Machine learning insights concerning inflammatory and liver-related risk comorbidities in non-communicable and viral diseases. World J Gastroenterol 2022; 28:6230-6248. [PMID: 36504554 PMCID: PMC9730439 DOI: 10.3748/wjg.v28.i44.6230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/07/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
The liver is a key organ involved in a wide range of functions, whose damage can lead to chronic liver disease (CLD). CLD accounts for more than two million deaths worldwide, becoming a social and economic burden for most countries. Among the different factors that can cause CLD, alcohol abuse, viruses, drug treatments, and unhealthy dietary patterns top the list. These conditions prompt and perpetuate an inflammatory environment and oxidative stress imbalance that favor the development of hepatic fibrogenesis. High stages of fibrosis can eventually lead to cirrhosis or hepatocellular carcinoma (HCC). Despite the advances achieved in this field, new approaches are needed for the prevention, diagnosis, treatment, and prognosis of CLD. In this context, the scientific com-munity is using machine learning (ML) algorithms to integrate and process vast amounts of data with unprecedented performance. ML techniques allow the integration of anthropometric, genetic, clinical, biochemical, dietary, lifestyle and omics data, giving new insights to tackle CLD and bringing personalized medicine a step closer. This review summarizes the investigations where ML techniques have been applied to study new approaches that could be used in inflammatory-related, hepatitis viruses-induced, and coronavirus disease 2019-induced liver damage and enlighten the factors involved in CLD development.
Collapse
Affiliation(s)
- J Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, Madrid Institute of Advanced Studies-Food Institute, Madrid 28049, Spain
| | - Marta Alonso-Bernáldez
- Precision Nutrition and Cardiometabolic Health, Madrid Institute of Advanced Studies-Food Institute, Madrid 28049, Spain
| | | | - Juan A Vargas-Nuñez
- Servicio de Medicina Interna, Hospital Universitario Puerta de Hierro Majadahonda, Madrid 28222, Majadahonda, Spain
| | - Ana Ramírez de Molina
- Molecular Oncology and Nutritional Genomics of Cancer, Madrid Institute of Advanced Studies-Food Institute, Madrid 28049, Spain
| | - Alberto Dávalos
- Laboratory of Epigenetics of Lipid Metabolism, Madrid Institute of Advanced Studies-Food Institute, Madrid 28049, Spain
| | - Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana 22390, Baja California, Mexico
| |
Collapse
|
57
|
Icaritin, a metabolite of Icarrin, Alleviates non-alcoholic fatty liver disease via inhibition of lipogenesis and ER stress. Eur J Pharmacol 2022; 937:175378. [DOI: 10.1016/j.ejphar.2022.175378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/13/2022]
|
58
|
Anapali M, Kaya-Dagistanli F, Akdemir AS, Aydemir D, Ulusu NN, Ulutin T, Uysal O, Tanriverdi G, Ozturk M. Combined resveratrol and vitamin D treatment ameliorate inflammation-related liver fibrosis, ER stress, and apoptosis in a high-fructose diet/streptozotocin-induced T2DM model. Histochem Cell Biol 2022; 158:279-296. [PMID: 35849204 DOI: 10.1007/s00418-022-02131-y] [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] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
A high fructose diet is a major cause of diabetes and various metabolic disorders, including fatty liver. In this study, we investigated the effects of resveratrol and vitamin D (VitD) treatments on endoplasmic reticulum (ER) stress, oxidative stress, inflammation, apoptosis, and liver regeneration in a rat model of type 2 diabetes mellitus, namely, T2DM Sprague-Dawley rats. This T2DM rat model was created through a combination treatment of a 10% fructose diet and 40 mg/kg streptozotocin (STZ). Resveratrol (1 mg/kg/day) and VitD (170/IU/week) were administered alone and in combination to both the diabetic and control groups. Immunohistochemical staining was performed to evaluate PCNA, NF-κB, TNF-α, IL-6, IL-1β, GRP78, and active caspase-3 in liver tissue. The TUNEL method and Sirius red staining were used to determine apoptosis and fibrosis, respectively. G6PD, 6-PGD, GR, and GST activities were measured to determine oxidative stress status. We found that the expressions of cytokines (TNF-α, IL-6, and IL-1β) correlated with NF-κB activation and were significantly increased in the T2DM rats. Increased GRP78 expression, indicating ER stress, increased in apoptotic cells, enhanced caspase-3 activation, and collagen accumulation surrounding the central vein were observed in the T2DM group compared with the other groups. The combination VitD + resveratrol treatment improved antioxidant defense via increasing G6PD, 6-PGD, GR, and GST activities compared to the diabetic groups. We concluded that the combined administration of resveratrol with VitD ameliorates the adverse effects of T2DM by regulating blood glucose levels, increasing antioxidant defense mechanisms, controlling ER stress, enhancing tissue regeneration, improving inflammation, and reducing apoptosis in liver cells. In conclusion, this study indicates that the combination treatment of resveratrol + VitD can be a beneficial option for preventing liver damage in fructose-induced T2DM.
Collapse
Affiliation(s)
- Merve Anapali
- Department of Medical Biology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Fatma Kaya-Dagistanli
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| | - Ayse Seda Akdemir
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Duygu Aydemir
- Department of Medical Biochemistry, School of Medicine, Koc University, Istanbul, Turkey.,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Nuriye Nuray Ulusu
- Department of Medical Biochemistry, School of Medicine, Koc University, Istanbul, Turkey.,Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Turgut Ulutin
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Omer Uysal
- Department of Biostatistics, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Gamze Tanriverdi
- Department of Histology and Embryology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Melek Ozturk
- Department of Medical Biology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey.
| |
Collapse
|
59
|
Shao G, Liu Y, Lu L, Zhang G, Zhou W, Wu T, Wang L, Xu H, Ji G. The Pathogenesis of HCC Driven by NASH and the Preventive and Therapeutic Effects of Natural Products. Front Pharmacol 2022; 13:944088. [PMID: 35873545 PMCID: PMC9301043 DOI: 10.3389/fphar.2022.944088] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a clinical syndrome with pathological changes that are similar to those of alcoholic hepatitis without a history of excessive alcohol consumption. It is a specific form of nonalcoholic fatty liver disease (NAFLD) that is characterized by hepatocyte inflammation based on hepatocellular steatosis. Further exacerbation of NASH can lead to cirrhosis, which may then progress to hepatocellular carcinoma (HCC). There is a lack of specific and effective treatments for NASH and NASH-driven HCC, and the mechanisms of the progression of NASH to HCC are unclear. Therefore, there is a need to understand the pathogenesis and progression of these diseases to identify new therapeutic approaches. Currently, an increasing number of studies are focusing on the utility of natural products in NASH, which is likely to be a promising prospect for NASH. This paper reviews the possible mechanisms of the pathogenesis and progression of NASH and NASH-derived HCC, as well as the potential therapeutic role of natural products in NASH and NASH-derived HCC.
Collapse
Affiliation(s)
- Gaoxuan Shao
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Liu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangtao Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
60
|
Li S, Wu Y, Jiang H, Zhou F, Ben A, Wang R, Hua C. Chicory polysaccharides alleviate high-fat diet-induced non-alcoholic fatty liver disease via alteration of lipid metabolism- and inflammation-related gene expression. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.03.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
61
|
Yamaguchi T, Yoshida K, Murata M, Suwa K, Tsuneyama K, Matsuzaki K, Naganuma M. Smad3 Phospho-Isoform Signaling in Nonalcoholic Steatohepatitis. Int J Mol Sci 2022; 23:ijms23116270. [PMID: 35682957 PMCID: PMC9181097 DOI: 10.3390/ijms23116270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/28/2022] [Accepted: 05/29/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis with insulin resistance, oxidative stress, lipotoxicity, adipokine secretion by fat cells, endotoxins (lipopolysaccharides) released by gut microbiota, and endoplasmic reticulum stress. Together, these factors promote NAFLD progression from steatosis to nonalcoholic steatohepatitis (NASH), fibrosis, and eventually end-stage liver diseases in a proportion of cases. Hepatic fibrosis and carcinogenesis often progress together, sharing inflammatory pathways. However, NASH can lead to hepatocarcinogenesis with minimal inflammation or fibrosis. In such instances, insulin resistance, oxidative stress, and lipotoxicity can directly lead to liver carcinogenesis through genetic and epigenetic alterations. Transforming growth factor (TGF)-β signaling is implicated in hepatic fibrogenesis and carcinogenesis. TGF-β type I receptor (TβRI) and activated-Ras/c-Jun-N-terminal kinase (JNK) differentially phosphorylate the mediator Smad3 to create two phospho-isoforms: C-terminally phosphorylated Smad3 (pSmad3C) and linker-phosphorylated Smad3 (pSmad3L). TβRI/pSmad3C signaling terminates cell proliferation, while constitutive Ras activation and JNK-mediated pSmad3L promote hepatocyte proliferation and carcinogenesis. The pSmad3L signaling pathway also antagonizes cytostatic pSmad3C signaling. This review addresses TGF-β/Smad signaling in hepatic carcinogenesis complicating NASH. We also discuss Smad phospho-isoforms as biomarkers predicting HCC in NASH patients with or without cirrhosis.
Collapse
Affiliation(s)
- Takashi Yamaguchi
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
- Correspondence: ; Tel.: +81-72-804-0101; Fax: +81-72-804-2524
| | - Katsunori Yoshida
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Miki Murata
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Kanehiko Suwa
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Koichi Tsuneyama
- Department of Pathology & Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto, Tokushima 770-8503, Japan;
| | - Koichi Matsuzaki
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| | - Makoto Naganuma
- Department of Gastroenterology and Hepatology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; (K.Y.); (M.M.); (K.S.); (K.M.); (M.N.)
| |
Collapse
|
62
|
Lastun VL, Levet C, Freeman M. The mammalian rhomboid protein RHBDL4 protects against endoplasmic reticulum stress by regulating the morphology and distribution of ER sheets. J Biol Chem 2022; 298:101935. [PMID: 35436469 PMCID: PMC9136127 DOI: 10.1016/j.jbc.2022.101935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 11/16/2022] Open
Abstract
In metazoans, the architecture of the endoplasmic reticulum (ER) differs between cell types and undergoes major changes throughout the cell cycle and according to physiological needs. Although much is known about how the different ER morphologies are generated and maintained, especially ER tubules, how context-dependent changes in ER shape and distribution are regulated and the factors involved are less well characterized, as are the factors that contribute to the positioning of the ER within the cell. By overexpression and KO experiments, we show that the levels of RHBDL4, an ER-resident rhomboid protease, modulate the shape and distribution of the ER, especially during conditions that require rapid changes in the ER sheet distribution, such as ER stress. We demonstrate that RHBDL4 interacts with cytoskeleton-linking membrane protein 63 (CLIMP-63), a protein involved in ER sheet stabilization, as well as with the cytoskeleton. Furthermore, we found that mice lacking RHBDL4 are sensitive to ER stress and develop liver steatosis, a phenotype associated with unresolved ER stress. Taken together, these data suggest a new physiological role for RHBDL4 and also imply that this function does not require its enzymatic activity.
Collapse
|
63
|
Ji S, Sun J, Bian C, Huang X, Ji H. PKA/ATGL signaling pathway is involved in ER stress-mediated lipolysis in adipocytes of grass carp (Ctenopharyngodon idella). FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:683-691. [PMID: 35460470 DOI: 10.1007/s10695-021-01032-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/13/2021] [Indexed: 06/14/2023]
Abstract
The relationship between endoplasmic reticulum stress (ER stress) and lipolysis in mammals has been widely studied, but it is relatively scarce in fish. The present study used grass carp Ctenopharyngodon idella as a model to investigate the effect of ER stress on lipolysis in adipocytes of fish. We found that ER stress evoked by tunicamycin (TM) treatment significantly induced lipolysis in adipocytes. Subsequently, in order to further investigate whether protein kinase A (PKA) is involved in ER stress-induced lipolysis, we treated adipocytes with PKA activator forskolin and inhibitor H89. The results showed that the mechanism was related to the activation of PKA, especially the catalytic subunit PRKACBa. Notably, we also found that PKA regulates lipolysis by targeting mRNA level and protein and enzyme activities of adipotriglyceride lipase (ATGL). Taken together, our findings suggest that PKA/ATGL signaling pathway is involved in ER stress-mediated lipolysis of grass carp adipocytes. It provides a theoretical basis for further study on the mechanism of lipolysis in fish and other vertebrates.
Collapse
Affiliation(s)
- Shanghong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Jian Sun
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Chenchen Bian
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Xiaocheng Huang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling, 712100, China.
| |
Collapse
|
64
|
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
|
65
|
Arulkumar R, Jung HJ, Noh SG, Chung HY. Soyasapogenol C from Fermented Soybean ( Glycine Max) Acting as a Novel AMPK/PPARα Dual Activator Ameliorates Hepatic Steatosis: A Novel SANDA Methodology. Int J Mol Sci 2022; 23:5468. [PMID: 35628280 PMCID: PMC9141180 DOI: 10.3390/ijms23105468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Soyasapogenol C (SSC), a derivative of soyasapogenol B (SSB), is specifically found high in many fermented soybean (Glycine max) products, including Cheonggukjang (in Korean). However, the biological activities for preventing and treating hepatic steatosis, and the precise underlying mechanisms of SSC, remain to be explored. (2) Methods: A novel SANDA (structural screening, ADMET prediction, network pharmacology, docking validation, and activity evaluation) methodology was used to examine whether SSC exerts hepatoprotective effects in silico and in vitro. (3) Results: SSC had better ADMET characteristics and a higher binding affinity with predicted targets chosen from network pathway analysis than SSB. SSC induced the phosphorylation of AMP-activated protein kinase (AMPK) and stimulated the nuclear translocation of peroxisome proliferator-activated receptor alpha (PPARα), further enhancing PPAR response element (PPRE) binding activity in HepG2 cells. Concurrently, SSC significantly inhibited triglyceride accumulation, which was associated with the suppression of lipogenesis genes and the enhancement of fatty acid oxidation gene expression in HepG2 cells. (4) Conclusions: Soyasapogenol C, discovered using a novel SANDA methodology from fermented soybean, is a novel AMPK/PPARα dual activator that is effective against hepatic steatosis. Dietary supplementation with soyasapogenol C may prevent the development of hepatic steatosis and other diseases associated with fat accumulation in the liver.
Collapse
Affiliation(s)
- Radha Arulkumar
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, Busan 46241, Korea; (R.A.); (S.G.N.)
| | - Hee Jin Jung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea;
| | - Sang Gyun Noh
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, Busan 46241, Korea; (R.A.); (S.G.N.)
| | - Hae Young Chung
- Interdisciplinary Research Program of Bioinformatics and Longevity Science, Pusan National University, Busan 46241, Korea; (R.A.); (S.G.N.)
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
66
|
Kim GT, Devi S, Sharma A, Cho KH, Kim SJ, Kim BR, Kwon SH, Park TS. Upregulation of the serine palmitoyltransferase subunit SPTLC2 by endoplasmic reticulum stress inhibits the hepatic insulin response. Exp Mol Med 2022; 54:573-584. [PMID: 35513574 PMCID: PMC9166747 DOI: 10.1038/s12276-022-00766-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/13/2021] [Accepted: 12/30/2021] [Indexed: 11/27/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is induced by various conditions, such as inflammation and the presence of excess nutrients. Abnormal accumulation of unfolded proteins leads to the activation of a collective signaling cascade, termed the unfolded protein response (UPR). ER stress is reported to perturb hepatic insulin response metabolism while promoting insulin resistance. Here, we report that ER stress regulates the de novo biosynthesis of sphingolipids via the activation of serine palmitoyltransferase (SPT), a rate-limiting enzyme involved in the de novo biosynthesis of ceramides. We found that the expression levels of Sptlc1 and Sptlc2, the major SPT subunits, were upregulated and that the cellular concentrations of ceramide and dihydroceramide were elevated by acute ER stress inducers in primary hepatocytes and HepG2 cells. Sptlc2 was upregulated and ceramide levels were elevated by tunicamycin in the livers of C57BL/6J wild-type mice. Analysis of the Sptlc2 promoter demonstrated that the transcriptional activation of Sptlc2 was mediated by the spliced form of X-box binding protein 1 (sXBP1). Liver-specific Sptlc2 transgenic mice exhibited increased ceramide levels in the liver and elevated fasting glucose levels. The insulin response was reduced by the inhibition of the phosphorylation of insulin receptor β (IRβ). Collectively, these results demonstrate that ER stress induces activation of the de novo biosynthesis of ceramide and contributes to the progression of hepatic insulin resistance via the reduced phosphorylation of IRβ in hepatocytes. A lipid molecule called ceramide is key to regulating the body’s insulin response, which controls blood sugar, and thus may hold keys to new treatments for metabolic diseases such as diabetes. Although ceramide levels were known to be raised in obesity and diabetes, the mechanism remained unclear. Tae-Sik Park at Gachon University, Sungnam, South Korea, and Sang-Ho Kwon at Augusta University, USA, and co-workers investigated how excess ceramide production is triggered and the blood sugar regulation consequences. They found that the liver-specific SPTLC2 transgenic mice fed a high-fat diet had increased levels of an enzyme activity of serine palmitoyltransferase which led to synthesis of high levels of ceramide in the liver. The high ceramide levels suppressed insulin signaling, imbalancing blood sugar levels and causing liver toxicity. Therapies that inhibit ceramide synthesis show promise for treatment of metabolic diseases.
Collapse
Affiliation(s)
- Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Shivani Devi
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Amitesh Sharma
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Kyung-Hee Cho
- Department of Life Science, Gachon University, Sungnam, Korea
| | - Su-Jung Kim
- Biomedical Research Center, Asan Institute for Life Sciences, Seoul, Korea
| | - Bo-Rahm Kim
- The Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam, Korea. .,Lipidomia Inc., Sungnam, Korea.
| |
Collapse
|
67
|
Prasad M, Jayaraman S, Rajagopal P, Veeraraghavan VP, Kumar PK, Piramanayagam S, Pari L. Diosgenin inhibits ER stress-induced inflammation in aorta via iRhom2/TACE mediated signaling in experimental diabetic rats: An in vivo and in silico approach. Chem Biol Interact 2022; 358:109885. [PMID: 35305976 DOI: 10.1016/j.cbi.2022.109885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 02/14/2022] [Accepted: 03/07/2022] [Indexed: 11/03/2022]
|
68
|
Flessa C, Kyrou I, Nasiri‐Ansari N, Kaltsas G, Kassi E, Randeva HS. Endoplasmic reticulum stress in nonalcoholic (metabolic associated) fatty liver disease (NAFLD/MAFLD). J Cell Biochem 2022; 123:1585-1606. [PMID: 35490371 DOI: 10.1002/jcb.30247] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023]
Affiliation(s)
- Christina‐Maria Flessa
- Department of Biological Chemistry, Medical School National and Kapodistrian University of Athens Athens Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) University Hospitals Coventry and Warwickshire NHS Trust Coventry UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) University Hospitals Coventry and Warwickshire NHS Trust Coventry UK
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School University of Warwick Coventry UK
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing Coventry University Coventry UK
- Aston Medical School, College of Health and Life Sciences Aston University Birmingham UK
- Department of Food Science & Human Nutrition Agricultural University of Athens Athens Greece
| | - Narjes Nasiri‐Ansari
- Department of Biological Chemistry, Medical School National and Kapodistrian University of Athens Athens Greece
| | - Gregory Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital National and Kapodistrian University of Athens Athens Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School National and Kapodistrian University of Athens Athens Greece
- Endocrine Unit, 1st Department of Propaedeutic and Internal Medicine, Laiko Hospital National and Kapodistrian University of Athens Athens Greece
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM) University Hospitals Coventry and Warwickshire NHS Trust Coventry UK
- Division of Translational and Experimental Medicine, Metabolic and Vascular Health, Warwick Medical School University of Warwick Coventry UK
| |
Collapse
|
69
|
Li Y, Huang S, Wang J, Dai J, Cai J, Yan S, Huang Z, He S, Wang P, Liu J, Liu Y. Phosphorylation at Ser 724 of the ER stress sensor IRE1α governs its activation state and limits ER stress-induced hepatosteatosis. J Biol Chem 2022; 298:101997. [PMID: 35500653 PMCID: PMC9144033 DOI: 10.1016/j.jbc.2022.101997] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/20/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Inositol-requiring enzyme 1 (IRE1) is an evolutionarily conserved sensor of endoplasmic reticulum (ER) stress and mediates a key branch of the unfolded protein response in eukaryotic cells. It is an ER-resident transmembrane protein that possesses Ser/Thr protein kinase and endoribonuclease (RNase) activities in its cytoplasmic region. IRE1 is activated through dimerization/oligomerization and autophosphorylation at multiple sites, acting through its RNase activity to restore the functional capacity of the ER. However, it remains poorly defined in vivo how the autophosphorylation events of endogenous IRE1 govern its dynamic activation and functional output. Here, we generated a mouse model harboring a S724A knock-in mutation (Ern1S724A/S724A) and investigated the importance of phosphorylation at Ser724 within the kinase activation loop of murine IRE1α. We found that in mouse embryonic fibroblast cells and in primary hepatocytes, S724A mutation resulted in markedly reduced IRE1α autophosphorylation in parallel with blunted activation of its RNase activity to catalyze X-box binding protein 1 (Xbp1) mRNA splicing. Furthermore, ablation of IRE1α phosphorylation at Ser724 exacerbated ER stress–induced hepatic steatosis in tunicamycin-treated Ern1S724A/S724A mice. This was accompanied by significantly decreased hepatic production of spliced XBP1 protein but increased CCAAT-enhancer–binding protein homologous protein (CHOP) level, along with suppressed expression of key metabolic regulators of fatty acid β-oxidation and lipid secretion. These results demonstrate a critical role of phosphorylation at Ser724 of IRE1α in dynamically controlling its kinase activity, and thus its autophosphorylation state, which is coupled to activation of its RNase activity in counteracting hepatic steatosis under ER stress conditions.
Collapse
Affiliation(s)
- Yang Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Frontier Science Center for Immunology and Metabolism; and the Institute for Advanced Studies; Wuhan University, Wuhan, China
| | - Shijia Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Frontier Science Center for Immunology and Metabolism; and the Institute for Advanced Studies; Wuhan University, Wuhan, China
| | - Jingsi Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Frontier Science Center for Immunology and Metabolism; and the Institute for Advanced Studies; Wuhan University, Wuhan, China
| | - Jianli Dai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China, China
| | - Jie Cai
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Frontier Science Center for Immunology and Metabolism; and the Institute for Advanced Studies; Wuhan University, Wuhan, China
| | - Shuai Yan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Zhiliang Huang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Shengqi He
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Frontier Science Center for Immunology and Metabolism; and the Institute for Advanced Studies; Wuhan University, Wuhan, China
| | - Ping Wang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jianmiao Liu
- Cellular Signaling Laboratory, Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences; Frontier Science Center for Immunology and Metabolism; and the Institute for Advanced Studies; Wuhan University, Wuhan, China.
| |
Collapse
|
70
|
Wang X, Xin H, Xing M, Gu X, Hao Y. Acute Endoplasmic Reticulum Stress Induces Inflammation Reaction, Complement System Activation, and Lipid Metabolism Disorder of Piglet Livers: A Proteomic Approach. Front Physiol 2022; 13:857853. [PMID: 35492579 PMCID: PMC9043290 DOI: 10.3389/fphys.2022.857853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022] Open
Abstract
Endoplasmic reticulum stress (ERS) is closely associated with the occurrence and development of many liver diseases. ERS models mostly include experimental animals such as rats and mice. However, pigs are more similar to humans with regards to digestion and metabolism, especially liver construction, yet few reports on ERS in pigs exist. In order to explore changes in the liver under ERS, we used tunicamycin (TM), which can cause liver jaundice and damage liver function, to establish acute ERS models in piglets using a low TM dosage (LD, 0.1 mg/kg body weight (bw)), high TM dosage (HD, 0.3 mg/kg bw), or vehicle for 48 h. We found that both LD- and HD-induced ERS, as verified by the ERS-linked proteins. Furthermore, the concentrations of the proinflammatory cytokines, namely, TNF-α and IL-6 were elevated in TM-treated piglet livers, and the plasma levels of IL-6 and CRP were also higher, indicating the occurrence of inflammation in TM-treated piglets. The complement system was activated in TM-treated piglets, as indicated by increased levels of complement factors and activation products C3, C5a, and AP50. In order to gain insights into the global changes in liver proteins under ERS, we performed an isobaric tag for relative and absolute quantitation (iTRAQ)-based proteomic analysis on the livers of HD- and vehicle-treated piglets. Proteomic analysis identified 311 differentially expressed proteins (DEPs) between the two groups, and a Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis suggested that the DEPs were mainly enriched in signaling pathways such as metabolic pathways, protein processing in the endoplasmic reticulum, and complement and coagulation cascades. Many proteins involved in protein folding, lipid transport, and oxidation were upregulated. Proteins involved in lipid synthesis were downregulated to alleviate liver steatosis, and most complement factors were upregulated to protect the body, and Pearson correlation analysis found that most of the DEPs in the complement and coagulation pathway were significantly correlated with plasma CRP, IL6 and AP50. Our results revealed that TM can activate ERS, marked by liver injury and steatosis, inflammatory reactions, and complement activation in piglets.
Collapse
Affiliation(s)
| | | | | | | | - Yue Hao
- *Correspondence: Xianhong Gu, ; Yue Hao,
| |
Collapse
|
71
|
Subramanian P, Gargani S, Palladini A, Chatzimike M, Grzybek M, Peitzsch M, Papanastasiou AD, Pyrina I, Ntafis V, Gercken B, Lesche M, Petzold A, Sinha A, Nati M, Thangapandi VR, Kourtzelis I, Andreadou M, Witt A, Dahl A, Burkhardt R, Haase R, Domingues AMDJ, Henry I, Zamboni N, Mirtschink P, Chung KJ, Hampe J, Coskun Ü, Kontoyiannis DL, Chavakis T. The RNA binding protein human antigen R is a gatekeeper of liver homeostasis. Hepatology 2022; 75:881-897. [PMID: 34519101 DOI: 10.1002/hep.32153] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS NAFLD is initiated by steatosis and can progress through fibrosis and cirrhosis to HCC. The RNA binding protein human antigen R (HuR) controls RNAs at the posttranscriptional level; hepatocyte HuR has been implicated in the regulation of diet-induced hepatic steatosis. The present study aimed to understand the role of hepatocyte HuR in NAFLD development and progression to fibrosis and HCC. APPROACH AND RESULTS Hepatocyte-specific, HuR-deficient mice and control HuR-sufficient mice were fed either a normal diet or an NAFLD-inducing diet. Hepatic lipid accumulation, inflammation, fibrosis, and HCC development were studied by histology, flow cytometry, quantitative PCR, and RNA sequencing. The liver lipidome was characterized by lipidomics analysis, and the HuR-RNA interactions in the liver were mapped by RNA immunoprecipitation sequencing. Hepatocyte-specific, HuR-deficient mice displayed spontaneous hepatic steatosis and fibrosis predisposition compared to control HuR-sufficient mice. On an NAFLD-inducing diet, hepatocyte-specific HuR deficiency resulted in exacerbated inflammation, fibrosis, and HCC-like tumor development. A multi-omic approach, including lipidomics, transcriptomics, and RNA immunoprecipitation sequencing revealed that HuR orchestrates a protective network of hepatic-metabolic and lipid homeostasis-maintaining pathways. Consistently, HuR-deficient livers accumulated, already at steady state, a triglyceride signature resembling that of NAFLD livers. Moreover, up-regulation of secreted phosphoprotein 1 expression mediated, at least partially, fibrosis development in hepatocyte-specific HuR deficiency on an NAFLD-inducing diet, as shown by experiments using antibody blockade of osteopontin. CONCLUSIONS HuR is a gatekeeper of liver homeostasis, preventing NAFLD-related fibrosis and HCC, suggesting that the HuR-dependent network could be exploited therapeutically.
Collapse
Affiliation(s)
- Pallavi Subramanian
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Sofia Gargani
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Alessandra Palladini
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Margarita Chatzimike
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Michal Grzybek
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Anastasios D Papanastasiou
- Department of Biomedical SciencesUniversity of West AtticaAthensGreece.,Histopathology UnitBiomedical Sciences Research Center "Alexander Fleming"VariGreece
| | - Iryna Pyrina
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Vasileios Ntafis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Bettina Gercken
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Mathias Lesche
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Andreas Petzold
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Anupam Sinha
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Marina Nati
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Veera Raghavan Thangapandi
- Department of Internal Medicine IUniversity Hospital and Faculty of Medicine, Technische Universität DresdenDresdenGermany
| | - Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany.,National Center for Tumor DiseasesPartner Site Dresden, Dresden and German Cancer Research CenterHeidelbergGermany.,York Biomedical Research Institute, Hull York Medical SchoolUniversity of YorkYorkUK
| | - Margarita Andreadou
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece
| | - Anke Witt
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Andreas Dahl
- DRESDEN-concept Genome CenterCenter for Molecular and Cellular BioengineeringTechnische Universität DresdenDresdenGermany
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory MedicineUniversity Hospital RegensburgRegensburgGermany
| | - Robert Haase
- Scientific Computing FacilityMax Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | | | - Ian Henry
- Scientific Computing FacilityMax Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Nicola Zamboni
- Institute of Molecular Systems BiologyETH ZurichZurichSwitzerland
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany
| | - Jochen Hampe
- Department of Internal Medicine IUniversity Hospital and Faculty of Medicine, Technische Universität DresdenDresdenGermany
| | - Ünal Coskun
- Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany
| | - Dimitris L Kontoyiannis
- Institute for Fundamental Biomedical Research (IFBR), Biomedical Sciences Research Centre "Alexander Fleming"VariGreece.,Department of Genetics, Development & Molecular Biology, School of BiologyAristotle University of ThessalonikiThessalonikiGreece
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory MedicineFaculty of MedicineTechnische Universität DresdenDresdenGermany.,Paul Langerhans Institute Dresden, Helmholtz Zentrum München, University Hospital and Faculty of MedicineTechnische Universität DresdenDresdenGermany.,German Center for Diabetes ResearchNeuherbergGermany.,National Center for Tumor DiseasesPartner Site Dresden, Dresden and German Cancer Research CenterHeidelbergGermany
| |
Collapse
|
72
|
Zhang DG, Xu XJ, Pantopoulos K, Zhao T, Zheng H, Luo Z. HSF1-SELENOS pathway mediated dietary inorganic Se-induced lipogenesis via the up-regulation of PPARγ expression in yellow catfish. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194802. [PMID: 35248747 DOI: 10.1016/j.bbagrm.2022.194802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023]
Abstract
At present, studies involved in the effects of dietary Se sources on lipid metabolism were very scarce and the underlying mechanism remains unknown. Previous studies reported that dietary Se sources differentially affected selenoprotein S (SELENOS) expression and SELENOS affected lipid metabolism via the inositol-requiring enzyme 1α (IRE1α)- spliced X-box binding protein 1 (XBP1s) pathway. Thus, we used yellow catfish as an experimental model to explore whether dietary selenium sources affected the hepatic lipid metabolism, and further determined the role of SELENOS-IRE1α-XBP1s pathway in dietary selenium sources affecting hepatic lipid metabolism. Compared with the selenomethionine (S-M) group, sodium selenite (SS) group possessed higher liver triglycerides (TGs) (34.7%), lipogenic enzyme activities (57.9-70.6%), and lower antioxidant enzyme activities (23.3-35.5%), increased protein levels of heat shock transcription factor 1 (HSF1) and SELENOS (1.17-fold and 47.4%, respectively), and XBP1s- peroxisome proliferators-activated receptor γ (PPARγ) pathway. Blocking SELENOS and PPARγ by RNA interference demonstrated that the SELENOS/XBP1s/PPARγ axis was critical for S-S-induced lipid accumulation. Moreover, S-S-induced upregulation of SELENOS was via the increased DNA binding capacity of HSF1 to SELENOS promoter, which activated the XBP1s/PPARγ pathway and promoted lipogenesis and lipid accumulation. XBP1s is required for S-S-induced upregulation of PPARγ expression. Our finding elucidated the mechanism of dietary Se sources affecting the lipid metabolism in the liver of yellow catfish and demonstrated novel function of SELENOS in metabolic regulation. Our study also suggested that seleno-methionine was a better Se source than selenite against abnormal lipid deposition in the liver of yellow catfish.
Collapse
Affiliation(s)
- Dian-Guang Zhang
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Xiao-Jian Xu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal H3T 1E2, Quebec, Canada
| | - Tao Zhao
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Hua Zheng
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
73
|
Malekpour-Dehkordi Z, Nourbakhsh M, Shahidi M, Sarraf N, Sharifi R. "Silymarin diminishes oleic acid-induced lipid accumulation in HepG2 cells by modulating the expression of endoplasmic reticulum stress markers". J Herb Med 2022. [DOI: 10.1016/j.hermed.2022.100565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
74
|
HAN S. Aging and gender-related effects of tauroursodeoxycholic acid treatment on liver functions, plasma lipid profile, and oxidative stress. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1023909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
75
|
Shu Z, Zhang G, Zhu X, Xiong W. Estrogen receptor α mediated M1/M2 macrophages polarization plays a critical role in NASH of female mice. Biochem Biophys Res Commun 2022; 596:63-70. [PMID: 35114586 DOI: 10.1016/j.bbrc.2022.01.085] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/23/2022] [Indexed: 12/11/2022]
Abstract
Owing to lacking protective effect of estrogen, OVX mice have higher risk of non-alcoholic fatty liver disease compared with normal female mice, when fed with high fat diet. Our study was to explore how estrogen protect against nonalcoholic steatohepatitis in female mice. We found that, lacking estrogen, M1 macrphages was activated and promoted steatohepatitis in obese OVX mice. And, ERα was responsible for estrogen to inhibit M1 macrphages activation and steatohepatitis. ERα knockdown aggravated M1 macrophages infiltration by transcriptionally upregulated its CCR2 expression. CCR2 antagonist effectively improved nonalcoholic steatohepatitis, ER stress and insulin resistance in ERα knockdown obese female mice. These results demonstrated ERα mediated M1 macrophages activation played a key role in nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Zhiping Shu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Guopeng Zhang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.
| | - Wenqian Xiong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, China.
| |
Collapse
|
76
|
Ward CP, Peng L, Yuen S, Halstead J, Palacios H, Nyangau E, Mohammed H, Ziari N, Dandan M, Frakes AE, Gildea HK, Dillin A, Hellerstein M. Aging alters the metabolic flux signature of the ER-unfolded protein response in vivo in mice. Aging Cell 2022; 21:e13558. [PMID: 35170180 PMCID: PMC8920450 DOI: 10.1111/acel.13558] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 11/16/2021] [Accepted: 12/25/2021] [Indexed: 01/23/2023] Open
Abstract
Age is a risk factor for numerous diseases, including neurodegenerative diseases, cancers, and diabetes. Loss of protein homeostasis is a central hallmark of aging. Activation of the endoplasmic reticulum unfolded protein response (UPRER ) includes changes in protein translation and membrane lipid synthesis. Using stable isotope labeling, a flux "signature" of the UPRER in vivo in mouse liver was developed by inducing ER stress with tunicamycin and measuring rates of both proteome-wide translation and de novo lipogenesis. Several changes in protein synthesis across ontologies were noted with age, including a more dramatic suppression of translation under ER stress in aged mice as compared with young mice. Binding immunoglobulin protein (BiP) synthesis rates and mRNA levels were increased more in aged than young mice. De novo lipogenesis rates decreased under ER stress conditions in aged mice, including both triglyceride and phospholipid fractions. In young mice, a significant reduction was seen only in the triglyceride fraction. These data indicate that aged mice have an exaggerated metabolic flux response to ER stress, which may indicate that aging renders the UPRER less effective in resolving proteotoxic stress.
Collapse
Affiliation(s)
- Catherine P. Ward
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Lucy Peng
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Samuel Yuen
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - John Halstead
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Hector Palacios
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Edna Nyangau
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Hussein Mohammed
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Naveed Ziari
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Mohamad Dandan
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Ashley E. Frakes
- Department of Molecular and Cellular BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Holly K. Gildea
- Department of Molecular and Cellular BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Andrew Dillin
- Department of Molecular and Cellular BiologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and ToxicologyUniversity of CaliforniaBerkeleyCaliforniaUSA
| |
Collapse
|
77
|
Pressly JD, Gurumani MZ, Varona Santos JT, Fornoni A, Merscher S, Al-Ali H. Adaptive and maladaptive roles of lipid droplets in health and disease. Am J Physiol Cell Physiol 2022; 322:C468-C481. [PMID: 35108119 PMCID: PMC8917915 DOI: 10.1152/ajpcell.00239.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Advances in the understanding of lipid droplet biology have revealed essential roles for these organelles in mediating proper cellular homeostasis and stress response. Lipid droplets were initially thought to play a passive role in energy storage. However, recent studies demonstrate that they have substantially broader functions, including protection from reactive oxygen species, endoplasmic reticulum stress, and lipotoxicity. Dysregulation of lipid droplet homeostasis is associated with various pathologies spanning neurological, metabolic, cardiovascular, oncological, and renal diseases. This review provides an overview of the current understanding of lipid droplet biology in both health and disease.
Collapse
Affiliation(s)
- Jeffrey D. Pressly
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Margaret Z. Gurumani
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Javier T. Varona Santos
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Alessia Fornoni
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Sandra Merscher
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Hassan Al-Ali
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida,3Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, Florida,4The Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, Florida,5Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
78
|
Tang D, Zhang Q, Duan H, Ye X, Liu J, Peng W, Wu C. Polydatin: A Critical Promising Natural Agent for Liver Protection via Antioxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9218738. [PMID: 35186191 PMCID: PMC8853764 DOI: 10.1155/2022/9218738] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022]
Abstract
Polydatin, one of the natural active small molecules, was commonly applied in protecting and treating liver disorders in preclinical studies. Oxidative stress plays vital roles in liver injury caused by various factors, such as alcohol, viral infections, dietary components, drugs, and other chemical reagents. It is reported that oxidative stress might be one of the main reasons in the progressive development of alcohol liver diseases (ALDs), nonalcoholic liver diseases (NAFLDs), liver injury, fibrosis, hepatic failure (HF), and hepatocellular carcinoma (HCC). In this paper, we comprehensively summarized the pharmacological effects and potential molecular mechanisms of polydatin for protecting and treating liver disorders via regulation of oxidative stress. According to the previous studies, polydatin is a versatile natural compound and exerts significantly protective and curative effects on oxidative stress-associated liver diseases via various molecular mechanisms, including amelioration of liver function and insulin resistance, inhibition of proinflammatory cytokines, lipid accumulation, endoplasmic reticulum stress and autophagy, regulation of PI3K/Akt/mTOR, and activation of hepatic stellate cells (HSCs), as well as increase of antioxidant enzymes (such as catalase (CAT), glutathione peroxidase (GPx), glutathione (GSH), superoxide dismutase (SOD), glutathione reductase (GR), and heme oxygenase-1 (HO-1)). In addition, polydatin acts as a free radical scavenger against reactive oxygen species (ROS) by its phenolic and ethylenic bond structure. However, further clinical investigations are still needed to explore the comprehensive molecular mechanisms and confirm the clinical treatment effect of polydatin in liver diseases related to regulation of oxidative stress.
Collapse
Affiliation(s)
- Dandan Tang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Qing Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Huxinyue Duan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Xun Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Jia Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Wei Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, No. 1166, Liutai Avenue, Chengdu 611137, China
| |
Collapse
|
79
|
Madhavan A, Kok BP, Rius B, Grandjean JMD, Alabi A, Albert V, Sukiasyan A, Powers ET, Galmozzi A, Saez E, Wiseman RL. Pharmacologic IRE1/XBP1s activation promotes systemic adaptive remodeling in obesity. Nat Commun 2022; 13:608. [PMID: 35105890 PMCID: PMC8807832 DOI: 10.1038/s41467-022-28271-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/18/2022] [Indexed: 01/08/2023] Open
Abstract
In obesity, signaling through the IRE1 arm of the unfolded protein response exerts both protective and harmful effects. Overexpression of the IRE1-regulated transcription factor XBP1s in liver or fat protects against obesity-linked metabolic deterioration. However, hyperactivation of IRE1 engages regulated IRE1-dependent decay (RIDD) and TRAF2/JNK pro-inflammatory signaling, which accelerate metabolic dysfunction. These pathologic IRE1-regulated processes have hindered efforts to pharmacologically harness the protective benefits of IRE1/XBP1s signaling in obesity-linked conditions. Here, we report the effects of a XBP1s-selective pharmacological IRE1 activator, IXA4, in diet-induced obese (DIO) mice. IXA4 transiently activates protective IRE1/XBP1s signaling in liver without inducing RIDD or TRAF2/JNK signaling. IXA4 treatment improves systemic glucose metabolism and liver insulin action through IRE1-dependent remodeling of the hepatic transcriptome that reduces glucose production and steatosis. IXA4-stimulated IRE1 activation also enhances pancreatic function. Our findings indicate that systemic, transient activation of IRE1/XBP1s signaling engenders multi-tissue benefits that integrate to mitigate obesity-driven metabolic dysfunction.
Collapse
Affiliation(s)
- Aparajita Madhavan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Bernard P Kok
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Bibiana Rius
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Julia M D Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Adekunle Alabi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Verena Albert
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ara Sukiasyan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Evan T Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Andrea Galmozzi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Department of Medicine, University of Wisconsin, Madison, WI, 53705, USA
| | - Enrique Saez
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
80
|
Zhang X, Huang C, Yuan Y, Jin S, Zhao J, Zhang W, Liang H, Chen X, Zhang B. FOXM1-mediated activation of phospholipase D1 promotes lipid droplet accumulation and reduces ROS to support paclitaxel resistance in metastatic cancer cells. Free Radic Biol Med 2022; 179:213-228. [PMID: 34808333 DOI: 10.1016/j.freeradbiomed.2021.11.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022]
Abstract
Chemoresistance is a major challenge for the treatment of cancer with metastasis. We investigated the mechanisms of lipid metabolites involved in drug resistance. Here, metastatic cancer cells isolated from mouse models were resistant to paclitaxel treatment in vitro and in vivo when compared with parental cancer cells. FOXM1, an oncogenic transcriptional factor, was highly expressed in metastatic cancer cells, and overexpression of FOXM1 conferred parental cancer cells resistance to paclitaxel. Lipidomic analysis showed that FOXM1 increased unsaturated triglyceride (TG) and phosphatidylcholine (PC) abundance, which are the main components of lipid droplet (LD). Inhibition of LD formation sensitized cells to paclitaxel. Mechanistically, the enzyme phospholipase D1 (PLD1) was identified as a potential effector target of FOXM1. PLD1 promoted LD accumulation, which reduced the level of reactive oxygen species (ROS) and maintained endoplasmic reticulum (ER) homeostasis in resistant cells with the treatment of paclitaxel. Moreover, inhibition of PLD1 reversed FOXM1-conferred paclitaxel resistance in vitro and in vivo. This study, for the first time, reveals the role of FOXM1-mediated PLD1 in LD accumulation and paclitaxel resistance. Targeting PLD1 or LD formation may help reverse chemoresistance in metastatic cancer cells. Generally, our results identified FOXM1 as a driver of paclitaxel resistance via activation of PLD1 to promote of LD accumulation, which contributes to the maintenace of ER homeostasis when metastatic cancer cells are confronted with ROS induced by paclitaxel.
Collapse
Affiliation(s)
- Xin Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sanshan Jin
- Department of Traditional Chinese Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Disease, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Key Laboratory of Hepato-Pancreato-Biliary Disease, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, 430030, China.
| |
Collapse
|
81
|
Duwaerts CC, Maiers JL. ER Disposal Pathways in Chronic Liver Disease: Protective, Pathogenic, and Potential Therapeutic Targets. Front Mol Biosci 2022; 8:804097. [PMID: 35174209 PMCID: PMC8841999 DOI: 10.3389/fmolb.2021.804097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum is a central player in liver pathophysiology. Chronic injury to the ER through increased lipid content, alcohol metabolism, or accumulation of misfolded proteins causes ER stress, dysregulated hepatocyte function, inflammation, and worsened disease pathogenesis. A key adaptation of the ER to resolve stress is the removal of excess or misfolded proteins. Degradation of intra-luminal or ER membrane proteins occurs through distinct mechanisms that include ER-associated Degradation (ERAD) and ER-to-lysosome-associated degradation (ERLAD), which includes macro-ER-phagy, micro-ER-phagy, and Atg8/LC-3-dependent vesicular delivery. All three of these processes are critical for removing misfolded or unfolded protein aggregates, and re-establishing ER homeostasis following expansion/stress, which is critical for liver function and adaptation to injury. Despite playing a key role in resolving ER stress, the contribution of these degradative processes to liver physiology and pathophysiology is understudied. Analysis of publicly available datasets from diseased livers revealed that numerous genes involved in ER-related degradative pathways are dysregulated; however, their roles and regulation in disease progression are not well defined. Here we discuss the dynamic regulation of ER-related protein disposal pathways in chronic liver disease and cell-type specific roles, as well as potentially targetable mechanisms for treatment of chronic liver disease.
Collapse
Affiliation(s)
- Caroline C. Duwaerts
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jessica L. Maiers
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
82
|
Ward CP, Peng L, Yuen S, Chang M, Karapetyan R, Nyangau E, Mohammed H, Palacios H, Ziari N, Joe LK, Frakes AE, Dandan M, Dillin A, Hellerstein MK. ER Unfolded Protein Response in Liver In Vivo Is Characterized by Reduced, Not Increased, De Novo Lipogenesis and Cholesterol Synthesis Rates with Uptake of Fatty Acids from Adipose Tissue: Integrated Gene Expression, Translation Rates and Metabolic Fluxes. Int J Mol Sci 2022; 23:ijms23031073. [PMID: 35162995 PMCID: PMC8835023 DOI: 10.3390/ijms23031073] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023] Open
Abstract
The unfolded protein response in the endoplasmic reticulum (UPRER) is involved in a number of metabolic diseases. Here, we characterize UPRER-induced metabolic changes in mouse livers in vivo through metabolic labeling and mass spectrometric analysis of lipid and proteome-wide fluxes. We induced UPRER by tunicamycin administration and measured synthesis rates of proteins, fatty acids and cholesterol, as well as RNA-seq. Contrary to reports in isolated cells, hepatic de novo lipogenesis and cholesterogenesis were markedly reduced, as were mRNA levels and synthesis rates of lipogenic proteins. H&E staining showed enrichment with lipid droplets while electron microscopy revealed ER morphological changes. Interestingly, the pre-labeling of adipose tissue prior to UPRER induction resulted in the redistribution of labeled fatty acids from adipose tissue to the liver, with replacement by unlabeled glycerol in the liver acylglycerides, indicating that the liver uptake was of free fatty acids, not whole glycerolipids. The redistribution of adipose fatty acids to the liver was not explicable by altered plasma insulin, increased fatty acid levels (lipolysis) or by reduced food intake. Synthesis of most liver proteins was suppressed under UPRER conditions, with the exception of BiP, other chaperones, protein disulfide isomerases, and proteins of ribosomal biogenesis. Protein synthesis rates generally, but not always, paralleled changes in mRNA. In summary, this combined approach, linking static changes with fluxes, revealed an integrated reduction of lipid and cholesterol synthesis pathways, from gene expression to translation and metabolic flux rates, under UPRER conditions. The reduced lipogenesis does not parallel human fatty liver disease. This approach provides powerful tools to characterize metabolic processes underlying hepatic UPRER in vivo.
Collapse
Affiliation(s)
- Catherine P. Ward
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Lucy Peng
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Samuel Yuen
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Michael Chang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Rozalina Karapetyan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Edna Nyangau
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Hussein Mohammed
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Hector Palacios
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Naveed Ziari
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Larry K. Joe
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 92093, USA; (L.K.J.); (A.E.F.); (A.D.)
| | - Ashley E. Frakes
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 92093, USA; (L.K.J.); (A.E.F.); (A.D.)
| | - Mohamad Dandan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
| | - Andrew Dillin
- Department of Molecular and Cellular Biology, University of California, Berkeley, CA 92093, USA; (L.K.J.); (A.E.F.); (A.D.)
| | - Marc K. Hellerstein
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 92093, USA; (C.P.W.); (L.P.); (S.Y.); (M.C.); (R.K.); (E.N.); (H.M.); (H.P.); (N.Z.); (M.D.)
- Correspondence:
| |
Collapse
|
83
|
Solomon TP, Carter S, Haus JM, Karstoft K, von Holstein-Rathlou S, Nielsen MS, Gillum MP. Plasma FGF21 concentrations are regulated by glucose independently of insulin and GLP-1 in lean, healthy humans. PeerJ 2022; 10:e12755. [PMID: 35111398 PMCID: PMC8783558 DOI: 10.7717/peerj.12755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 12/15/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) treatment improves metabolic homeostasis in diverse species, including humans. Physiologically, plasma FGF21 levels increase modestly after glucose ingestion, but it is unclear whether this is mediated by glucose itself or due to a secondary effect of postprandial endocrine responses. A refined understanding of the mechanisms that control FGF21 release in humans may accelerate the development of small-molecule FGF21 secretagogues to treat metabolic disease. This study aimed to determine whether FGF21 secretion is stimulated by elevations in plasma glucose, insulin, or glucagon-like peptide-1 (GLP-1) in humans. METHODS Three groups of ten healthy participants were included in a parallel-group observational study. Group A underwent a hyperglycemic infusion; Group B underwent a 40 mU/m2/min hyperinsulinemic euglycemic clamp; Group C underwent two pancreatic clamps (to suppress endogenous insulin secretion) with euglycemic and hyperglycemic stages with an infusion of either saline or 0.5 pmol/kg/min GLP-1. Plasma FGF21 concentrations were measured at baseline and during each clamp stage by ELISA. RESULTS Plasma FGF21 was unaltered during hyperglycemic infusion and hyperinsulinemic euglycemic clamps, compared to baseline. FGF21 was, however, increased by hyperglycemia under pancreatic clamp conditions (P < 0.05), while GLP-1 infusion under pancreatic clamp conditions did not change circulating FGF21 levels. CONCLUSION Increases in plasma FGF21 are likely driven directly by changes in plasma glucose independent of changes in insulin or GLP-1 secretion. Ecologically valid postprandial investigations are now needed to confirm our observations from basic science infusion models.
Collapse
Affiliation(s)
- Thomas P.J. Solomon
- School of Sport, Exercise and Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Edgbaston, United Kingdom,Institute of Metabolism and Systems Research, College of Medical & Dental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Steven Carter
- School of Sport, Exercise and Rehabilitation Sciences, College of Life & Environmental Sciences, University of Birmingham, Edgbaston, United Kingdom
| | - Jacob M. Haus
- School of Kinesiology, University of Michigan - Ann Arbor, Michigan, United States of America
| | - Kristian Karstoft
- Centre of Inflammation and Metabolism, Rigshospitalet, Copenhagen, Denmark,Centre for Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Stephanie von Holstein-Rathlou
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette S. Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthew P. Gillum
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
84
|
Phillips MA, Arnold KR, Vue Z, Beasley HK, Garza-Lopez E, Marshall AG, Morton DJ, McReynolds MR, Barter TT, Hinton A. Combining Metabolomics and Experimental Evolution Reveals Key Mechanisms Underlying Longevity Differences in Laboratory Evolved Drosophila melanogaster Populations. Int J Mol Sci 2022; 23:1067. [PMID: 35162994 PMCID: PMC8835531 DOI: 10.3390/ijms23031067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/22/2022] Open
Abstract
Experimental evolution with Drosophila melanogaster has been used extensively for decades to study aging and longevity. In recent years, the addition of DNA and RNA sequencing to this framework has allowed researchers to leverage the statistical power inherent to experimental evolution to study the genetic basis of longevity itself. Here, we incorporated metabolomic data into to this framework to generate even deeper insights into the physiological and genetic mechanisms underlying longevity differences in three groups of experimentally evolved D. melanogaster populations with different aging and longevity patterns. Our metabolomic analysis found that aging alters mitochondrial metabolism through increased consumption of NAD+ and increased usage of the TCA cycle. Combining our genomic and metabolomic data produced a list of biologically relevant candidate genes. Among these candidates, we found significant enrichment for genes and pathways associated with neurological development and function, and carbohydrate metabolism. While we do not explicitly find enrichment for aging canonical genes, neurological dysregulation and carbohydrate metabolism are both known to be associated with accelerated aging and reduced longevity. Taken together, our results provide plausible genetic mechanisms for what might be driving longevity differences in this experimental system. More broadly, our findings demonstrate the value of combining multiple types of omic data with experimental evolution when attempting to dissect mechanisms underlying complex and highly polygenic traits such as aging.
Collapse
Affiliation(s)
- Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR 97331, USA;
| | - Kenneth R. Arnold
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA 92697, USA; (K.R.A.); (T.T.B.)
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Z.V.); (H.K.B.); (A.G.M.)
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Z.V.); (H.K.B.); (A.G.M.)
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Edgar Garza-Lopez
- Hinton and Garza-Lopez Family Consulting Company, Iowa City, IA 52246, USA;
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Z.V.); (H.K.B.); (A.G.M.)
| | - Derrick J. Morton
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA;
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, Huck Institute of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA;
| | - Thomas T. Barter
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA 92697, USA; (K.R.A.); (T.T.B.)
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; (Z.V.); (H.K.B.); (A.G.M.)
- Hinton and Garza-Lopez Family Consulting Company, Iowa City, IA 52246, USA;
| |
Collapse
|
85
|
Quercetin Reduces Lipid Accumulation in a Cell Model of NAFLD by Inhibiting De Novo Fatty Acid Synthesis through the Acetyl-CoA Carboxylase 1/AMPK/PP2A Axis. Int J Mol Sci 2022; 23:ijms23031044. [PMID: 35162967 PMCID: PMC8834998 DOI: 10.3390/ijms23031044] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/06/2023] Open
Abstract
Dysregulation of de novo lipogenesis (DNL) has recently gained strong attention as being one of the critical factors that contribute to the assessment of non-alcoholic fatty liver disease (NAFLD). NAFLD is often diagnosed in patients with dyslipidemias and type 2 diabetes; thus, an interesting correlation can be deduced between high hematic free fatty acids and glucose excess in the DNL dysregulation. In the present study, we report that, in a cellular model of NAFLD, the coexistence of elevated glucose and FFA conditions caused the highest cellular lipid accumulation. Deepening the molecular mechanisms of the DNL dysregulation—RT-qPCR and immunoblot analysis demonstrated increased expression of mitochondrial citrate carrier (CiC), cytosolic acetyl-CoA carboxylase 1 (ACACA), and diacylglycerol acyltransferase 2 (DGAT2) involved in fatty acids and triglycerides synthesis, respectively. XBP-1, an endoplasmic reticulum stress marker, and SREBP-1 were the transcription factors connected to the DNL activation. Quercetin (Que), a flavonoid with strong antioxidant properties, and noticeably reduced the lipid accumulation and the expression of SREBP-1 and XBP-1, as well as of their lipogenic gene targets in steatotic cells. The anti-lipogenic action of Que mainly occurs through a strong phosphorylation of ACACA, which catalyzes the committing step in the DNL pathway. The high level of ACACA phosphorylation in Que-treated cells was explained by the intervention of AMPK together with the reduction of enzymatic activity of PP2A phosphatase. Overall, our findings highlight a direct anti-lipogenic effect of Que exerted through inhibition of the DNL pathway by acting on ACACA/AMPK/PP2A axis; thus, suggesting this flavonoid as a promising molecule for the NAFLD treatment.
Collapse
|
86
|
Zhang Q, Liu X, Piao C, Jiao Z, Ma Y, Wang Y, Liu T, Xu J, Wang H. Effect of conditioned medium from adipose derived mesenchymal stem cells on endoplasmic reticulum stress and lipid metabolism after hepatic ischemia reperfusion injury and hepatectomy in swine. Life Sci 2022; 289:120212. [PMID: 34896163 DOI: 10.1016/j.lfs.2021.120212] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
AIMS Hepatic ischemia reperfusion injury (HIRI) is associated with liver failure after liver transplantation and hepatectomy. Thus, this study aims to explore the effect of conditioned medium from adipose derived stem cells (ADSC-CM) on endoplasmic reticulum stress (ERS) and lipid metabolism after HIRI combined with hepatectomy in miniature pigs. MAIN METHODS A model of HIRI combined with hepatectomy in miniature pigs was established. The expression of ERS-related proteins and lipid metabolism related genes, as well as triglyceride (TG), total cholesterol (TC), high density lipoprotein (HDL), very low density lipoprotein (VLDL) and acetyl-CoA carboxylase 1 (ACC1) level were measured in liver tissues. KEY FINDINGS Both ADSCs and ADSC-CM could improve the damage in the ultrastructure of hepatocytes. ADSC-CM significantly decreased the protein expression of GRP78, ATF6, XBP1, p-eIF2α, ATF4, p-JNK and CHOP. Oil red O staining revealed that the degree of hepatocyte steatosis was also significantly reduced after treatment with ADSC-CM. In addition, ADSC-CM remarkably decreased TG, TC, HDL and ACC1 level in liver tissues, while enhanced VLDL content. Finally, SREBP1, SCAP, FASN, ACC1, HMGCR and HMGCS1 mRNA expression was also markedly downregulated in liver tissues. SIGNIFICANCE Injection of ADSC-CM into the hepatic parenchymal could represent a novel cell-free therapeutic approach to improve HIRI combined with hepatectomy injury. The inhibition of ERS and the improvement of lipid metabolism in the hepatocytes might be a potential mechanism used by ADSC-CM to prevent liver injury from HIRI combined with hepatectomy.
Collapse
Affiliation(s)
- Qianzhen Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, PR China
| | - Xiaoning Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Chenxi Piao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Zhihui Jiao
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, PR China
| | - Yajun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yue Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Tao Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiayuan Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Hongbin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
87
|
Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity 2022; 55:31-55. [PMID: 35021057 PMCID: PMC8773457 DOI: 10.1016/j.immuni.2021.12.013] [Citation(s) in RCA: 897] [Impact Index Per Article: 299.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 01/13/2023]
Abstract
Obesity leads to chronic, systemic inflammation and can lead to insulin resistance (IR), β-cell dysfunction, and ultimately type 2 diabetes (T2D). This chronic inflammatory state contributes to long-term complications of diabetes, including non-alcoholic fatty liver disease (NAFLD), retinopathy, cardiovascular disease, and nephropathy, and may underlie the association of type 2 diabetes with other conditions such as Alzheimer's disease, polycystic ovarian syndrome, gout, and rheumatoid arthritis. Here, we review the current understanding of the mechanisms underlying inflammation in obesity, T2D, and related disorders. We discuss how chronic tissue inflammation results in IR, impaired insulin secretion, glucose intolerance, and T2D and review the effect of inflammation on diabetic complications and on the relationship between T2D and other pathologies. In this context, we discuss current therapeutic options for the treatment of metabolic disease, advances in the clinic and the potential of immune-modulatory approaches.
Collapse
Affiliation(s)
- Theresa V. Rohm
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel T. Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland
| | - Jerrold M. Olefsky
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, CH-4031 Basel, Switzerland.,Department of Biomedicine (DBM), University of Basel, University Hospital Basel, CH-4031 Basel, Switzerland.,Correspondence:
| |
Collapse
|
88
|
Maiorana A, Lepri FR, Novelli A, Dionisi-Vici C. Hypoglycaemia Metabolic Gene Panel Testing. Front Endocrinol (Lausanne) 2022; 13:826167. [PMID: 35422763 PMCID: PMC9001947 DOI: 10.3389/fendo.2022.826167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 12/31/2022] Open
Abstract
A large number of inborn errors of metabolism present with hypoglycemia. Impairment of glucose homeostasis may arise from different biochemical pathways involving insulin secretion, fatty acid oxidation, ketone bodies formation and degradation, glycogen metabolism, fructose and galactose metabolism, branched chain aminoacids and tyrosine metabolism, mitochondrial function and glycosylation proteins mechanisms. Historically, genetic analysis consisted of highly detailed molecular testing of nominated single genes. However, more recently, the genetic heterogeneity of these conditions imposed to perform extensive molecular testing within a useful timeframe via new generation sequencing technology. Indeed, the establishment of a rapid diagnosis drives specific nutritional and medical therapies. The biochemical and clinical phenotypes are critical to guide the molecular analysis toward those clusters of genes involved in specific pathways, and address data interpretation regarding the finding of possible disease-causing variants at first reported as variants of uncertain significance in known genes or the discovery of new disease genes. Also, the trio's analysis allows genetic counseling for recurrence risk in further pregnancies. Besides, this approach is allowing to expand the phenotypic characterization of a disease when pathogenic variants give raise to unexpected clinical pictures. Multidisciplinary input and collaboration are increasingly key for addressing the analysis and interpreting the significance of the genetic results, allowing rapidly their translation from bench to bedside.
Collapse
Affiliation(s)
- Arianna Maiorana
- Division of Metabolism, Department of Pediatrics Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), Rome, Italy
- *Correspondence: Arianna Maiorana,
| | - Francesca Romana Lepri
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unity, Ospedale Pediatrico Bambino Gesù, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), Rome, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unity, Ospedale Pediatrico Bambino Gesù, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), Rome, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolism, Department of Pediatrics Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico), Rome, Italy
| |
Collapse
|
89
|
Parthasarathy G, Malhi H. Assessment of Lipotoxic Endoplasmic Reticulum (ER) Stress in Nonalcoholic Steatohepatitis (NASH). Methods Mol Biol 2022; 2455:243-254. [PMID: 35212999 PMCID: PMC9333415 DOI: 10.1007/978-1-0716-2128-8_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hepatocyte lipotoxicity is a hallmark of nonalcoholic steatohepatitis (NASH), and lipid induced liver injury occurs, in part, via activation of endoplasmic reticulum (ER) stress. Consequently, the unfolded protein response (UPR) is initiated, driven by three key ER transmembrane proteins, resulting in downstream responses that are dynamic and interconnected. Thus, careful interrogation of these pathways is required to investigate the complex role of ER stress in NASH. Herein, we describe different mechanisms of, and in vitro assays for assessment of lipotoxic ER stress in mouse hepatocytes.
Collapse
Affiliation(s)
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
90
|
Riera-Heredia N, Lutfi E, Balbuena-Pecino S, Vélez EJ, Dias K, Beaumatin F, Gutiérrez J, Seiliez I, Capilla E, Navarro I. The autophagy response during adipogenesis of primary cultured rainbow trout (Oncorhynchus mykiss) adipocytes. Comp Biochem Physiol B Biochem Mol Biol 2021; 258:110700. [PMID: 34848371 DOI: 10.1016/j.cbpb.2021.110700] [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] [Received: 08/30/2021] [Revised: 11/18/2021] [Accepted: 11/25/2021] [Indexed: 10/19/2022]
Abstract
Adipogenesis is a tightly regulated process, and the involvement of autophagy has been recently proposed in mammalian models. In rainbow trout, two well-defined phases describe the development of primary cultured adipocyte cells: proliferation and differentiation. Nevertheless, information on the transcriptional profile at the onset of differentiation and the potential role of autophagy in this process is scarce. In the present study, the cells showed an early and transient induction of several adipogenic transcription factors genes' expression (i.e., cebpa and cebpb) along with the morphological changes (round shape filled with small lipid droplets) typical of the onset of adipogenesis. Then, the expression of various lipid metabolism-related genes involving the synthesis (fas), uptake (fatp1 and cd36), accumulation (plin2) and mobilization (hsl) of lipids, characteristic of the mature adipocyte, increased. In parallel, several autophagy markers (i.e., atg4b, gabarapl1 and lc3b) mirrored the expression of those adipogenic-related genes, suggesting a role of autophagy during in vitro fish adipogenesis. In this regard, the incubation of preadipocytes with lysosomal inhibitors (Bafilomycin A1 or Chloroquine), described to prevent autophagy flux, delayed the process of adipogenesis (i.e., cell remodelling), thus suggesting a possible relationship between autophagy and adipocyte differentiation in trout. Moreover, the disruption of the autophagic flux altered the expression of some key adipogenic genes such as cebpa and pparg. Overall, this study contributes to improve our knowledge on the regulation of rainbow trout adipocyte differentiation, and highlights for the first time in fish the involvement of autophagy on adipogenesis, suggesting a close-fitting connection between both processes.
Collapse
Affiliation(s)
- Natàlia Riera-Heredia
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain
| | - Esmail Lutfi
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain
| | - Sara Balbuena-Pecino
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain
| | - Emilio J Vélez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain; Université de Pau et des Pays de l'Adour, E2S UPPA, INRAE, UMR1419, Nutrition Métabolisme et Aquaculture, F-64310 Saint-Pée-sur-Nivelle, France
| | - Karine Dias
- Université de Pau et des Pays de l'Adour, E2S UPPA, INRAE, UMR1419, Nutrition Métabolisme et Aquaculture, F-64310 Saint-Pée-sur-Nivelle, France
| | - Florian Beaumatin
- Université de Pau et des Pays de l'Adour, E2S UPPA, INRAE, UMR1419, Nutrition Métabolisme et Aquaculture, F-64310 Saint-Pée-sur-Nivelle, France
| | - Joaquim Gutiérrez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain
| | - Iban Seiliez
- Université de Pau et des Pays de l'Adour, E2S UPPA, INRAE, UMR1419, Nutrition Métabolisme et Aquaculture, F-64310 Saint-Pée-sur-Nivelle, France
| | - Encarnación Capilla
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain
| | - Isabel Navarro
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona 08028, Spain.
| |
Collapse
|
91
|
Ramdas Nair A, Lakhiani P, Zhang C, Macchi F, Sadler KC. A permissive epigenetic landscape facilitates distinct transcriptional signatures of activating transcription factor 6 in the liver. Genomics 2021; 114:107-124. [PMID: 34863900 DOI: 10.1016/j.ygeno.2021.11.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/31/2021] [Accepted: 11/26/2021] [Indexed: 12/01/2022]
Abstract
Restoring homeostasis following proteostatic stress hinges on a stress-specific transcriptional signature. How these signatures are regulated is unknown. We use functional genomics to uncover how activating transcription factor 6 (ATF6), a central factor in the unfolded protein response, regulates its target genes in response to toxicant induced and physiological stress in the liver. We identified 652 conserved putative ATF6 targets (CPATs), which functioned in metabolism, development and proteostasis. Strikingly, Atf6 activation in the zebrafish liver by transgenic nAtf6 overexpression, ethanol and arsenic exposure resulted in a distinct CPAT signature for each; with only 34 CPATs differentially expressed in all conditions. In contrast, during liver regeneration in mice resulted in a dynamic differential expression pattern of 53% of CPATs. These CPATs were distinguished by residing in open chromatin, H3K4me3 occupancy and the absence of H3K27me3 on their promoters. This suggests that a permissive epigenetic landscape allows stress-specific Atf6 target gene expression.
Collapse
Affiliation(s)
- Anjana Ramdas Nair
- Program in Biology, New York University Abu Dhabi, PO Box. 129188, Abu Dhabi, United Arab Emirates
| | - Priyanka Lakhiani
- Program in Biology, New York University Abu Dhabi, PO Box. 129188, Abu Dhabi, United Arab Emirates
| | - Chi Zhang
- Program in Biology, New York University Abu Dhabi, PO Box. 129188, Abu Dhabi, United Arab Emirates
| | - Filippo Macchi
- Program in Biology, New York University Abu Dhabi, PO Box. 129188, Abu Dhabi, United Arab Emirates
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, PO Box. 129188, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
92
|
Hello from the other side: Membrane contact of lipid droplets with other organelles and subsequent functional implications. Prog Lipid Res 2021; 85:101141. [PMID: 34793861 DOI: 10.1016/j.plipres.2021.101141] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Lipid droplets (LDs) are ubiquitous organelles that play crucial roles in response to physiological and environmental cues. The identification of several neutral lipid synthesizing and regulatory protein complexes have propelled significant advance on the mechanisms of LD biogenesis in the endoplasmic reticulum (ER). Increasing evidence suggests that distinct proteins and regulatory factors, which localize to membrane contact sites (MCS), are involved not only in interorganellar lipid exchange and transport, but also function in other important cellular processes, including autophagy, mitochondrial dynamics and inheritance, ion signaling and inter-regulation of these MCS. More and more tethers and molecular determinants are associated to MCS and to a diversity of cellular and pathophysiological processes, demonstrating the dynamics and importance of these junctions in health and disease. The conjugation of lipids with proteins in supramolecular complexes is known to be paramount for many biological processes, namely membrane biosynthesis, cell homeostasis, regulation of organelle division and biogenesis, and cell growth. Ultimately, this physical organization allows the contact sites to function as crucial metabolic hubs that control the occurrence of chemical reactions. This leads to biochemical and metabolite compartmentalization for the purposes of energetic efficiency and cellular homeostasis. In this review, we will focus on the structural and functional aspects of LD-organelle interactions and how they ensure signaling exchange and metabolites transfer between organelles.
Collapse
|
93
|
Moreira JLDS, Barbosa SMB, Gonçalves Júnior J. Pathophysiology and molecular mechanisms of liver injury in severe forms of COVID-19: An integrative review. Clin Res Hepatol Gastroenterol 2021; 45:101752. [PMID: 34303828 PMCID: PMC8299216 DOI: 10.1016/j.clinre.2021.101752] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS SARS-CoV-2 has primary pulmonary impairment, but other organs such as the liver can also be affected. This implies a worsening of patient's prognosis and an increase in morbidity and mortality. The metabolic pathways and molecular factors involved in the genesis of this injury are still unknown. Therefore, we aimed to carry out an integrative review about the pathophysiology and possible molecular mechanisms of liver injury by COVID-19. METHODS We carried out an integrative literature review in the following databases: PubMed, Scopus, and Embase from December 2020 to March 2021 using the following descriptors: # 1 "COVID-19" (MeSH) AND / OR # 2 "Liver injury" (MeSH) AND / OR # 3 "Pathophysiology" (MesH). RESULTS The data were extracted and divided into two main themes, for heuristic purposes: "Hepatotropism and SARS-CoV-2", and "Pathophysiological hypotheses for liver injury associated with SARS-CoV-2". CONCLUSIONS The virus seems to promote liver damage through five mechanisms: direct injury, humoral and cellular inflammatory response, hypoxemia caused by a decrease in the effective circulating volume, reinfection through the portal system, and use of drugs in the treatment. The literature also points out that the expression of the angiotensin-converting enzyme II and transmembrane serine protease 2 receptors is expressive in cholangiocyte and is present in hepatocytes, which is a risk factor for the virus to enter these cells. Finally, patients with previous liver disease appear to be more susceptible to liver injury by COVID-19.
Collapse
Affiliation(s)
| | | | - Jucier Gonçalves Júnior
- Departament of Internal Medicine - Division of Rheumathology, Universidade de São Paulo (USP), São Paulo, Brazil.
| |
Collapse
|
94
|
Zhang Y, Lim CU, Sikirzhytski V, Naderi A, Chatzistamou I, Kiaris H. Propensity to endoplasmic reticulum stress in deer mouse fibroblasts predicts skin inflammation and body weight gain. Dis Model Mech 2021; 14:272498. [PMID: 34661243 PMCID: PMC8543066 DOI: 10.1242/dmm.049113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
The unfolded protein response (UPR) is involved in the pathogenesis of metabolic disorders, yet whether variations in the UPR among individuals influence the propensity for metabolic disease remains unexplored. Using outbred deer mice as a model, we show that the intensity of UPR in fibroblasts isolated early in life predicts the extent of body weight gain after high-fat diet (HFD) administration. Contrary to those with intense UPR, animals with moderate UPR in fibroblasts and therefore displaying compromised stress resolution did not gain body weight but developed inflammation, especially in the skin, after HFD administration. Fibroblasts emerged as potent modifiers of this differential responsiveness to HFD, as indicated by the comparison of the UPR profiles of fibroblasts responding to fatty acids in vitro, by correlation analyses between UPR and proinflammatory cytokine-associated transcriptomes, and by BiP (also known as HSPA5) immunolocalization in skin lesions from animals receiving HFD. These results suggest that the UPR operates as a modifier of an individual's propensity for body weight gain in a manner that, at least in part, involves the regulation of an inflammatory response by skin fibroblasts. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Youwen Zhang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Chang-Uk Lim
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Vitali Sikirzhytski
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Asieh Naderi
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA
| | - Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.,Peromyscus Genetic Stock Center, University of South Carolina, Columbia, SC 29208, USA
| |
Collapse
|
95
|
Zhang L, La X, Tian J, Li H, Li A, Liu Y, Wu C, Li Z. The phytochemical vitexin and syringic acid derived from foxtail fillet bran inhibit breast cancer cells proliferation via GRP78/SREBP-1/SCD1 signaling axis. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104620] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
96
|
Kern P, Balzer NR, Blank N, Cygon C, Wunderling K, Bender F, Frolov A, Sowa JP, Bonaguro L, Ulas T, Homrich M, Kiermaier E, Thiele C, Schultze JL, Canbay A, Bauer R, Mass E. Creld2 function during unfolded protein response is essential for liver metabolism homeostasis. FASEB J 2021; 35:e21939. [PMID: 34549824 DOI: 10.1096/fj.202002713rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022]
Abstract
The unfolded protein response (UPR) is associated with hepatic metabolic function, yet it is not well understood how endoplasmic reticulum (ER) disturbance might influence metabolic homeostasis. Here, we describe the physiological function of Cysteine-rich with EGF-like domains 2 (Creld2), previously characterized as a downstream target of the ER-stress signal transducer Atf6. To this end, we generated Creld2-deficient mice and induced UPR by injection of tunicamycin. Creld2 augments protein folding and creates an interlink between the UPR axes through its interaction with proteins involved in the cellular stress response. Thereby, Creld2 promotes tolerance to ER stress and recovery from acute stress. Creld2-deficiency leads to a dysregulated UPR and causes the development of hepatic steatosis during ER stress conditions. Moreover, Creld2-dependent enhancement of the UPR assists in the regulation of energy expenditure. Furthermore, we observed a sex dimorphism in human and mouse livers with only male patients showing an accumulation of CRELD2 protein during the progression from non-alcoholic fatty liver disease to non-alcoholic steatohepatitis and only male Creld2-deficient mice developing hepatic steatosis upon aging. These results reveal a Creld2 function at the intersection between UPR and metabolic homeostasis and suggest a mechanism in which chronic ER stress underlies fatty liver disease in males.
Collapse
Affiliation(s)
- Paul Kern
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Developmental Genetics & Molecular Physiology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Nora R Balzer
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Nelli Blank
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Cornelia Cygon
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Klaus Wunderling
- Biochemistry & Cell Biology of Lipids, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Franziska Bender
- Developmental Genetics & Molecular Physiology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Alex Frolov
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Jan-Peter Sowa
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany.,Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics at the Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), University of Bonn, Bonn, Germany
| | - Thomas Ulas
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics at the Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), University of Bonn, Bonn, Germany
| | - Mirka Homrich
- Immune and Tumor Biology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Eva Kiermaier
- Immune and Tumor Biology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Christoph Thiele
- Biochemistry & Cell Biology of Lipids, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics at the Deutsche Zentrum für Neurodegenerative Erkrankungen (DZNE), University of Bonn, Bonn, Germany.,Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Ali Canbay
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany.,Department of Medicine, Ruhr University Bochum, University Hospital Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Reinhard Bauer
- Developmental Genetics & Molecular Physiology, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Elvira Mass
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
97
|
Dong XC, Chowdhury K, Huang M, Kim HG. Signal Transduction and Molecular Regulation in Fatty Liver Disease. Antioxid Redox Signal 2021; 35:689-717. [PMID: 33906425 PMCID: PMC8558079 DOI: 10.1089/ars.2021.0076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Fatty liver disease is a major liver disorder in the modern societies. Comprehensive understanding of the pathophysiology and molecular mechanisms is essential for the prevention and treatment of the disease. Recent Advances: Remarkable progress has been made in the recent years in basic and translational research in the field of fatty liver disease. Multiple signaling pathways have been implicated in the development of fatty liver disease, including AMP-activated protein kinase, mechanistic target of rapamycin kinase, endoplasmic reticulum stress, oxidative stress, inflammation, transforming growth factor β, and yes1-associated transcriptional regulator/transcriptional coactivator with PDZ-binding motif (YAP/TAZ). In addition, critical molecular regulations at the transcriptional and epigenetic levels have been linked to the pathogenesis of fatty liver disease. Critical Issues: Some critical issues remain to be solved so that research findings can be translated into clinical applications. Robust and reliable biomarkers are needed for diagnosis of different stages of the fatty liver disease. Effective and safe molecular targets remain to be identified and validated. Prevention strategies require solid scientific evidence and population-wide feasibility. Future Directions: As more data are generated with time, integrative approaches are needed to comprehensively understand the disease pathophysiology and mechanisms at multiple levels from population, organismal system, organ/tissue, to cell. The interactions between genes and environmental factors require deeper investigation for the purposes of prevention and personalized treatment of fatty liver disease. Antioxid. Redox Signal. 35, 689-717.
Collapse
Affiliation(s)
- Xiaocheng Charlie Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of BioHealth Informatics, School of Informatics and Computing, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Kushan Chowdhury
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Hyeong Geug Kim
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
98
|
Kumar S, Duan Q, Wu R, Harris EN, Su Q. Pathophysiological communication between hepatocytes and non-parenchymal cells in liver injury from NAFLD to liver fibrosis. Adv Drug Deliv Rev 2021; 176:113869. [PMID: 34280515 PMCID: PMC11792083 DOI: 10.1016/j.addr.2021.113869] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/16/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a multifactorial disease that encompasses a spectrum of pathological conditions, ranging from simple steatosis (NAFL), nonalcoholic steatohepatitis (NASH), fibrosis/cirrhosis which can further progress to hepatocellular carcinoma and liver failure. The progression of NAFL to NASH and liver fibrosis is closely associated with a series of liver injury resulting from lipotoxicity, oxidative stress, redox imbalance (excessive nitric oxide), ER stress, inflammation and apoptosis that occur sequentially in different liver cells which ultimately leads to the activation of liver regeneration and fibrogenesis, augmenting collagen and extracellular matrix deposition and promoting liver fibrosis and cirrhosis. Type 2 diabetes is a significant risk factor in NAFLD development by accelerating liver damage. Here, we overview recent findings from human study and animal models on the pathophysiological communication among hepatocytes (HCs), Kupffer cells (KCs), hepatic stellate cells (HSCs) and liver sinusoidal endothelial cells (LSECs) during the disease development. The mechanisms of crucial signaling pathways, including Toll-like receptor, TGFβ and hedgehog mediated hepatic injury are also discussed. We further highlight the potentials of precisely targeting hepatic individual cell-type using nanotechnology as therapeutic strategy for the treatment of NASH and liver fibrosis.
Collapse
Affiliation(s)
- Santosh Kumar
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast BT9 5DL, United Kingdom
| | - Qihua Duan
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast BT9 5DL, United Kingdom
| | - Rongxue Wu
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Edward N Harris
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| | - Qiaozhu Su
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast BT9 5DL, United Kingdom.
| |
Collapse
|
99
|
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
|
100
|
Bramlage KS, Bhattacharjee J, Kirby M, Myronovych A, Gupta R, Gonzalez RMS, Xanthakos S, Bove K, Kohli R. A Diet High in Fat and Fructose Induces Early Hepatic Mitochondrial Aging. J Pediatr Gastroenterol Nutr 2021; 73:99-102. [PMID: 34135298 PMCID: PMC8549102 DOI: 10.1097/mpg.0000000000003068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT To investigate the effect of high fructose diet on ultrastructure and content of hepatic mitochondria, we randomized 6-8 weeks old male C57Bl6/J mice to ad lib chow or high-fat-high-fructose (HF2) diet for 32 weeks. HF2-fed mice gained more weight, had higher plasma alanine aminotransferase, and fasting glucose levels and increased hepatic triglyceride content at all time points compared to chow-fed mice. HF2-fed mice had lower mitochondrial to nuclear DNA ratio compared to chow-fed mice. HF2-fed mice had lower average mitochondrial surface area and the number of mitochondria compared to chow-fed mice. HF2-fed mice had higher expression of the hepatic endoplasmic reticulum stress marker Chop, compared to chow-fed mice. A diet high in fat and fructose leads to enhanced hepatic mitochondrial aging, depletion, and dysfunction, which may be important determinants of nonalcoholic steatohepatitis pathogenesis.
Collapse
Affiliation(s)
- Kristin S. Bramlage
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Jashdeep Bhattacharjee
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA
| | - Michelle Kirby
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Andriy Myronovych
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Rohun Gupta
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA
| | | | - Stavra Xanthakos
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kevin Bove
- Department of Pediatrics, Division of Pathology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital Los Angeles, Los Angeles, CA
| |
Collapse
|