1
|
Gargiulo S, Albanese S, Megna R, Gramanzini M, Marsella G, Vecchiarelli L. Veterinary medical care in rodent models of stroke: Pitfalls and refinements to balance quality of science and animal welfare. Neuroscience 2025; 572:269-302. [PMID: 39894435 DOI: 10.1016/j.neuroscience.2025.01.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/25/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
Rodent models of cerebral ischemia provide a valuable contribution to a better understanding of stroke pathophysiology, to validate diagnostic methods, and to enable testing of new treatments for ischemia-reperfusion damage and comorbidities. However, ethical concerns have led to increased attention to the welfare aspects of such models. Supportive therapies are an essential part of the overall animal care and use program and should be tailored to the experimental model being studied, the regulatory requirements, and research objectives to achieve high-quality preclinical studies and ethical research practices. On the other hand, the use of veterinary medical treatments in preclinical models of stroke must balance the needs of animal care and potential sources of bias in experimental results. This report provides a systematic review of the scientific literature covering the relevant period from years 1988 to September 2024, with the aim to investigating veterinary medical interventions useful to minimize suffering in rodent models of stroke without producing experimental bias. The research findings, consolidated from 181 selected studies, published from 1991 to 2023, indicate the feasibility of implementing personalized protocols of anesthesia, analgesics, antibiotics, and other supportive therapies in rodent models of stroke, while avoiding scientific interferences. These data fill a gap in current knowledge and could be of interest for an interdisciplinary audience working with rodent models of stroke, stimulating further refinements to safeguard both animal welfare and the validity of experimental findings, and may promote the culture of ethical conduct in various research fields and disciplines.
Collapse
Affiliation(s)
- Sara Gargiulo
- Institute of Clinical Physiology, National Research Council, Via Fiorentina 1, 53100 Siena, Italy.
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, National Research Council, 80131 Naples, Italy.
| | - Rosario Megna
- Institute of Biostructures and Bioimaging, National Research Council, 80131 Naples, Italy.
| | - Matteo Gramanzini
- Institute of Chemical Sciences and Technologies "Giulio Natta", National Research Council, L.go F. Vito, 00168 Rome, Italy.
| | - Gerardo Marsella
- Animal Care Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - Lidovina Vecchiarelli
- Animal Welfare at Animal and Plant Health Agency, Department for Environment Food and Rural Affairs, Midlands, UK.
| |
Collapse
|
2
|
Eccles-Miller JA, Johnson TD, Baldwin WS. Sexually Dimorphic Effects of CYP2B6 in the Development of Fasting-Mediated Steatosis in Mice: Role of the Oxylipin Products 9-HODE and 9-HOTrE. Biomedicines 2025; 13:295. [PMID: 40002708 PMCID: PMC11853041 DOI: 10.3390/biomedicines13020295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Cytochrome P450 2B6 (CYP2B6) is a sexually dimorphic, anti-obesity CYP enzyme responsible for the metabolism of xeno- and endobiotics, including the metabolism of polyunsaturated fatty acids (PUFAs) into 9-hydroxyoctadecadienoic acid (9-HODE) and 9-hydroxyoctadecatrienoic acid (9-HOTrE). However, humanized CYP2B6 transgenic (hCYP2B6-Tg) mice are sensitive to diet-induced hepatic steatosis despite their resistance to obesity. The purpose of this study was to determine if 9-HODE, 9-HOTrE, or other factors contribute to the sexually dimorphic steatosis observed in hCYP2B6-Tg mice. Results: Cyp2b9/10/13-null (Cyp2b-null) mice were injected with either 9-HODE or 9-HOTrE for 2 days and were then subjected to a fasting period of 20 h to induce steatosis. Serum lipids were moderately increased, especially in females, after 9-HODE (triglycerides (TGs), very low-density lipoproteins (VLDLs)) and 9-HOTrE (high-density lipoproteins (HDLs), low-density lipoproteins (LDLs), cholesterol) treatment. No change in hepatic lipids and few changes in hepatic gene expression were observed in mice treated with either oxylipin, suggesting that these oxylipins had minimal to moderate effects. Therefore, to further investigate CYP2B6's role in steatosis, hCYP2B6-Tg and Cyp2b-null mice were subjected to a 20 h fast and compared. Both male and female hCYP2B6-Tg mice exhibited increased steatosis compared to Cyp2b-null mice. Serum cholesterol, triglycerides, HDLs, and VLDLs were increased in hCYP2B6-Tg males. Serum triglycerides and VLDLs were decreased in hCYP2B6-Tg females, suggesting the greater hepatic retention of lipids in females. Hepatic oxylipin profiles revealed eight perturbed oxylipins in female hCYP2B6-Tg mice and only one in males when compared to Cyp2b-null mice. RNA-seq also demonstrated greater effects in females in terms of the number of genes and gene ontology (GO) terms perturbed. There were only a few overlapping GO terms between sexes, and lipid metabolic processes were enriched in hCYP2B6-Tg male mice but were repressed in hCYP2B6-Tg females compared to Cyp2b-nulls. Conclusions: hCYP2B6-Tg mice are sensitive to fasting-mediated steatosis in males and females, although the responses are different. In addition, the oxylipins 9-HODE and 9-HOTrE are unlikely to be the primary cause of CYP2B6's pro-steatotic effects.
Collapse
Affiliation(s)
| | | | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, SC 29634, USA; (J.A.E.-M.); (T.D.J.)
| |
Collapse
|
3
|
Chen J, Wang S, Yin X, Duan C, Li J, Liu YQ, Zhang Y. Relationship between rumen microbiota and pregnancy toxemia in ewes. Front Vet Sci 2024; 11:1472334. [PMID: 39397808 PMCID: PMC11466943 DOI: 10.3389/fvets.2024.1472334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Introduction Pregnancy toxemia (PT) is a nutritional metabolic disease of ewes in late pregnancy. This study aimed to reveal the relationship between rumen microbiota and PT. Methods We selected 10 healthy ewes (CON) and 10 pregnancy toxemia ewes (PT) at 135 days of gestation according to the blood β-hydroxybutyrate (BHBA), glucose (Glu) concentrations and clinical symptoms. Blood and rumen fluid were collected before morning feeding to determine serum biochemical indices and rumen fermentation parameters. Total DNA of rumen fluid was extracted and the V3-V4 regions of 16S rRNA were amplified by PCR for high-throughput sequencing. Results The results showed that the serum concentrations of Glu, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), uric acid (UA), creatinine (Cr), acetate, propionate, butyrate, and microbial crude protein (MCP) were decreased (p < 0.05) and the concentrations of BHBA, aspartate aminotransferase (AST), acetate to propionate ratio (A/P), and ammonia nitrogen (NH3-N)were higher (p < 0.05) in PT ewes than those in CON ewes. 16S rRNA analysis showed the differences of β-diversity were observed in rumen microbiota between CON and PT ewes. At the phylum level, the relative abundance of Bacteroidota and Proteobacteria were higher (p < 0.01), while Firmicutes was lower (p < 0.01) in PT ewes. At the genus level, the relative Prevotella, Butyrivibrio, Ruminococcus, Lachnospiraceae_AC2044_group, Lachnospiraceae_XPB1014_group, Lachnospiraceae_ND3007_group, and Oribacterium were lower (p < 0.01) in PT ewes. Meanwhile, the relative abundance of Oribacterium, Butyrivibrio, Ruminococcus, and Lachnospiraceae_AC2044_group were positively correlated (p < 0.01) with Glu, INS, acetate, propionate, and butyrate, and negatively correlated (p < 0.01) with BHBA, P, GC, AST, and A/P. Discussion In conclusion, the decrease of Oribacterium, Butyrivibrio, Ruminococcus, and Lachnospiraceae_AC2044_group in the rumen of PT ewes reduced the concentrations of volatile fatty acids (acetate, propionate, and butyrate) and serum Glu, and increased BHBA concentration, indicating that the differences in rumen bacteria genera were related to pregnancy toxemia of ewes.
Collapse
Affiliation(s)
- Jiaxin Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Siwei Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
- Hebei Key Laboratory of Crop Cultivation Physiology and Green Production, Institute of Cereal and Oil Crops, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, China
| | - Xuejiao Yin
- College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Chunhui Duan
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Jinhui Li
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yue-qin Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yingjie Zhang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
4
|
Huang X, Liu X, Li X, Zhang Y, Gao J, Yang Y, Jiang Y, Gao H, Sun C, Xuan L, Zhao L, Song J, Bao H, Zhou Z, Li S, Zhang X, Lu Y, Zhong X, Yang B, Pan Z. Cullin-associated and neddylation-dissociated protein 1 (CAND1) alleviates NAFLD by reducing ubiquitinated degradation of ACAA2. Nat Commun 2023; 14:4620. [PMID: 37528093 PMCID: PMC10394019 DOI: 10.1038/s41467-023-40327-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/24/2023] [Indexed: 08/03/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disorder with high morbidity and mortality. The current study aims to explore the role of Cullin-associated and neddylation-dissociated protein 1 (CAND1) in the development of NAFLD and the underlying mechanisms. CAND1 is reduced in the liver of NAFLD male patients and high fat diet (HFD)-fed male mice. CAND1 alleviates palmitate (PA) induced lipid accumulation in vitro. Hepatocyte-specific knockout of CAND1 exacerbates HFD-induced liver injury in HFD-fed male mice, while hepatocyte-specific knockin of CAND1 ameliorates these pathological changes. Mechanistically, deficiency of CAND1 enhances the assembly of Cullin1, F-box only protein 42 (FBXO42) and acetyl-CoA acyltransferase 2 (ACAA2) complexes, and thus promotes the ubiquitinated degradation of ACAA2. ACAA2 overexpression abolishes the exacerbated effects of CAND1 deficiency on NAFLD. Additionally, androgen receptor binds to the -187 to -2000 promoter region of CAND1. Collectively, CAND1 mitigates NAFLD by inhibiting Cullin1/FBXO42 mediated ACAA2 degradation.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Xin Liu
- The Department of Histology and Embryology, Harbin Medical University, Harbin, 150086, China
| | - Xingda Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (The Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Yang Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Jianjun Gao
- The Department of Hepatopancreatobility, Surgery Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ying Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Yuan Jiang
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Haiyu Gao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Chongsong Sun
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Lina Xuan
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Lexin Zhao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Jiahui Song
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Hairong Bao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Zhiwen Zhou
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Shangxuan Li
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Xiaofang Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China
| | - Yanjie Lu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China.
| | - Xiangyu Zhong
- The Department of Hepatopancreatobility, Surgery Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Baofeng Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019 Research Unit 070, Harbin, Heilongjiang, 150086, P. R. China.
- State Key Laboratory, Harbin Medical University, Harbin, 150086, China.
| | - Zhenwei Pan
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research. Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150086, P. R. China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019 Research Unit 070, Harbin, Heilongjiang, 150086, P. R. China.
- Key Laboratory of Cell Transplantation, The First Affiliated Hospital, Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
5
|
Vázquez-Borrego MC, Del Río-Moreno M, Pyatkov M, Sarmento-Cabral A, Mahmood M, Pelke N, Wnek M, Cordoba-Chacon J, Waxman DJ, Puchowicz MA, McGuinness OP, Kineman RD. Direct and systemic actions of growth hormone receptor (GHR)-signaling on hepatic glycolysis, de novo lipogenesis and insulin sensitivity, associated with steatosis. Metabolism 2023; 144:155589. [PMID: 37182789 PMCID: PMC10843389 DOI: 10.1016/j.metabol.2023.155589] [Citation(s) in RCA: 2] [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] [Received: 01/31/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Evidence is accumulating that growth hormone (GH) protects against the development of steatosis and progression of non-alcoholic fatty liver disease (NAFLD). GH may control steatosis indirectly by altering systemic insulin sensitivity and substrate delivery to the liver and/or by the direct actions of GH on hepatocyte function. APPROACH To better define the hepatocyte-specific role of GH receptor (GHR) signaling on regulating steatosis, we used a mouse model with adult-onset, hepatocyte-specific GHR knockdown (aHepGHRkd). To prevent the reduction in circulating insulin-like growth factor 1 (IGF1) and the subsequent increase in GH observed after aHepGHRkd, subsets of aHepGHRkd mice were treated with adeno-associated viral vectors (AAV) driving hepatocyte-specific expression of IGF1 or a constitutively active form of STAT5b (STAT5bCA). The impact of hepatocyte-specific modulation of GHR, IGF1 and STAT5b on carbohydrate and lipid metabolism was studied across multiple nutritional states and in the context of hyperinsulinemic:euglycemic clamps. RESULTS Chow-fed male aHepGHRkd mice developed steatosis associated with an increase in hepatic glucokinase (GCK) and ketohexokinase (KHK) expression and de novo lipogenesis (DNL) rate, in the post-absorptive state and in response to refeeding after an overnight fast. The aHepGHRkd-associated increase in hepatic KHK, but not GCK and steatosis, was dependent on hepatocyte expression of carbohydrate response element binding protein (ChREBP), in re-fed mice. Interestingly, under clamp conditions, aHepGHRkd also increased the rate of DNL and expression of GCK and KHK, but impaired insulin-mediated suppression of hepatic glucose production, without altering plasma NEFA levels. These effects were normalized with AAV-mediated hepatocyte expression of IGF1 or STAT5bCA. Comparison of the impact of AAV-mediated hepatocyte IGF1 versus STAT5bCA in aHepGHRkd mice across multiple nutritional states, indicated the restorative actions of IGF1 are indirect, by improving systemic insulin sensitivity, independent of changes in the liver transcriptome. In contrast, the actions of STAT5b are due to the combined effects of raising IGF1 and direct alterations in the hepatocyte gene program that may involve suppression of BCL6 and FOXO1 activity. However, the direct and IGF1-dependent actions of STAT5b cannot fully account for enhanced GCK activity and lipogenic gene expression observed after aHepGHRkd, suggesting other GHR-mediated signals are involved. CONCLUSION These studies demonstrate hepatocyte GHR-signaling controls hepatic glycolysis, DNL, steatosis and hepatic insulin sensitivity indirectly (via IGF1) and directly (via STAT5b). The relative contribution of these indirect and direct actions of GH on hepatocytes is modified by insulin and nutrient availability. These results improve our understanding of the physiologic actions of GH on regulating adult metabolism to protect against NAFLD progression.
Collapse
Affiliation(s)
- Mari C Vázquez-Borrego
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Mercedes Del Río-Moreno
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Maxim Pyatkov
- Department of Biology & Bioinformatics Program, Boston University, Boston, MA, United States of America
| | - André Sarmento-Cabral
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Mariyah Mahmood
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Natalie Pelke
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Magdalena Wnek
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - Jose Cordoba-Chacon
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America
| | - David J Waxman
- Department of Biology & Bioinformatics Program, Boston University, Boston, MA, United States of America
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States of America
| | - Rhonda D Kineman
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, United States of America; Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States of America.
| |
Collapse
|
6
|
Wang X, Liu SY, Hu GS, Wang HY, Zhang GL, Cen X, Xiang ST, Liu W, Li P, Ye H, Zhao TJ. DDB1 prepares brown adipocytes for cold-induced thermogenesis. LIFE METABOLISM 2022; 1:39-53. [PMID: 39872690 PMCID: PMC11749000 DOI: 10.1093/lifemeta/loac003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 01/30/2025]
Abstract
Brown adipose tissue (BAT) plays a key role in thermogenesis during acute cold exposure. However, it remains unclear how BAT is prepared to rapidly turn on thermogenic genes. Here, we show that damage-specific DNA binding protein 1 (DDB1) mediates the rapid transcription of thermogenic genes upon acute cold exposure. Adipose- or BAT-specific Ddb1 knockout mice show severely whitened BAT and significantly decreased expression of thermogenic genes. These mice develop hypothermia when subjected to acute cold exposure at 4 °C and partial lipodystrophy on a high-fat diet due to deficiency in fatty acid oxidation. Mechanistically, DDB1 binds the promoters of Ucp1 and Ppargc1a and recruits positive transcriptional elongation factor b (P-TEFb) to release promoter-proximally paused RNA polymerase II (Pol II), thereby enabling rapid and synchronized transcription of thermogenic genes upon acute cold exposure. Our findings have thus provided a regulatory mechanism of how BAT is prepared to respond to acute cold challenge.
Collapse
Affiliation(s)
- Xu Wang
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Qi Zhi Institute, Shanghai, China
| | - Shen-Ying Liu
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guo-Sheng Hu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, China
| | - Hao-Yan Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Guo-Liang Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiang Cen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Si-Ting Xiang
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen Liu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, China
| | - Peng Li
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Qi Zhi Institute, Shanghai, China
- State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Haobin Ye
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tong-Jin Zhao
- State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Qi Zhi Institute, Shanghai, China
| |
Collapse
|
7
|
Jain R, Wade G, Ong I, Chaurasia B, Simcox J. Determination of tissue contributions to the circulating lipid pool in cold exposure via systematic assessment of lipid profiles. J Lipid Res 2022; 63:100197. [PMID: 35300982 PMCID: PMC9234243 DOI: 10.1016/j.jlr.2022.100197] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 01/07/2023] Open
Abstract
Plasma lipid levels are altered in chronic conditions such as type 2 diabetes and cardiovascular disease as well as during acute stresses such as fasting and cold exposure. Advances in MS-based lipidomics have uncovered a complex plasma lipidome of more than 500 lipids that serve functional roles, including as energy substrates and signaling molecules. This plasma lipid pool is maintained through regulation of tissue production, secretion, and uptake. A major challenge in understanding the lipidome complexity is establishing the tissues of origin and uptake for various plasma lipids, which is valuable for determining lipid functions. Using cold exposure as an acute stress, we performed global lipidomics on plasma and in nine tissues that may contribute to the circulating lipid pool. We found that numerous species of plasma acylcarnitines (ACars) and ceramides (Cers) were significantly altered upon cold exposure. Through computational assessment, we identified the liver and brown adipose tissue as major contributors and consumers of circulating ACars, in agreement with our previous work. We further identified the kidney and intestine as novel contributors to the circulating ACar pool and validated these findings with gene expression analysis. Regression analysis also identified that the brown adipose tissue and kidney are interactors with the plasma Cer pool. Taken together, these studies provide an adaptable computational tool to assess tissue contribution to the plasma lipid pool. Our findings have further implications in understanding the function of plasma ACars and Cers, which are elevated in metabolic diseases.
Collapse
Affiliation(s)
- Raghav Jain
- Department of Biochemistry, University of Wisconsin-Madison, Wisconsin, USA
| | - Gina Wade
- Department of Biochemistry, University of Wisconsin-Madison, Wisconsin, USA
| | - Irene Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Wisconsin, USA
| | - Bhagirath Chaurasia
- Division of Endocrinology, Department of Internal Medicine, Carver College of Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa, USA
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Wisconsin, USA.
| |
Collapse
|
8
|
Mice blocking Ser347 phosphorylation of pregnane x receptor develop hepatic fasting-induced steatosis and hypertriglyceridemia. Biochem Biophys Res Commun 2022; 615:75-80. [PMID: 35609418 PMCID: PMC9233068 DOI: 10.1016/j.bbrc.2022.05.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 11/20/2022]
Abstract
Nuclear receptor Pregnane X Receptor (PXR; NR1I2) has transcriptional regulation functions for energy homeostasis in the liver. Mouse PXR has a conserved phosphorylation motif at serine 347 (serine 350 in humans) within the ligand-binding domain. PXR phosphorylated at this motif is expressed in mouse livers in response to fasting. Mice with a PXR∗Ser347Ala knockin mutation (PXR KI) were generated to block phosphorylation, and utilized to investigate the role of Ser347 phosphorylation in vivo. PXR KI mice had decreased body weight at 8-weeks of age and had much greater weight loss after fasting compared with PXR WT mice. The cDNA microarray analysis of hepatic mRNAs showed that cell death or apoptotic signaling was induced in fasting PXR KI mice. Moreover, increasing hepatic lipids, triglycerides and the development of hypertriglyceridemia were observed in fasting PXR KI mice. These findings are indicative that blocking phosphorylation prevents mice from maintaining hepatic energy homeostasis. Thus, phosphorylated PXR may be an essential factor to prevent the liver from developing damage caused by fasting.
Collapse
|
9
|
Wu G, Dai Y, Yan Y, Zheng X, Zhang H, Li H, Chen W. The lactate receptor GPR81 mediates hepatic lipid metabolism and the therapeutic effect of metformin on experimental NAFLDs. Eur J Pharmacol 2022; 924:174959. [PMID: 35430208 DOI: 10.1016/j.ejphar.2022.174959] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 01/06/2023]
Abstract
The lactate receptor G protein-coupled receptor 81 (GPR81) has been recently implicated in lipolysis in adipose tissue. In this study, we accidently discovered the role of GPR81 in hepatic lipid metabolism. Data clearly showed that hepatic GPR81 was markedly up-regulated in fasted mice, whereas it was severely down-regulated in obese mice. Genetic deficiency of GPR81 impaired ketogenic response, enhanced hepatic lipid accumulation, and exacerbated hepatosteatosis under acute fasting conditions. Mechanically, we demonstrated that hepatic GPR81 might function as a modulator of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), activate the downsream transcription of liver carnitine o-palmitoyltransferase 1(L-CPT1), and thereby control the influx of fatty acids into mitochondria for β-oxidation. Importantly, metformin improved experimental nonalcoholic fatty liver disease (NAFLDs) in a GPR81-dependent manner. Collectively, GPR81 was critical for hepatic lipid homeostasis and activation of hepatic GPR81 might represent a promising strategy for the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Guanglu Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yufeng Dai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Yongheng Yan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Xu Zheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
10
|
The monoacylglycerol acyltransferase pathway contributes to triacylglycerol synthesis in HepG2 cells. Sci Rep 2022; 12:4943. [PMID: 35322811 PMCID: PMC8943211 DOI: 10.1038/s41598-022-08946-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/14/2022] [Indexed: 12/19/2022] Open
Abstract
The monoacylglycerol acyltransferase (MGAT) pathway has a well-established role in the small intestine where it facilitates the absorption of dietary fat. In enterocytes, MGAT participates in the resynthesis of triacylglycerol using substrates (monoacylglycerol and fatty acids) generated in the gut lumen from the breakdown of triacylglycerol consumed in the diet. MGAT activity is also present in the liver, but its role in triacylglycerol metabolism in this tissue remains unclear. The predominant MGAT isoforms present in human liver appear to be MGAT2 and MGAT3. The objective of this study was to use selective small molecule inhibitors of MGAT2 and MGAT3 to determine the contributions of these enzymes to triacylglycerol production in liver cells. We found that pharmacological inhibition of either enzyme had no effect on TG mass in HepG2 cells but did alter lipid droplet size and number. Inhibition of MGAT2 did result in decreased DG and TG synthesis and TG secretion. Interestingly, MGAT2 preferentially utilized 2-monoacylglycerol derived from free glycerol and not from exogenously added 2-monoacylglycerol. In contrast, inhibition of MGAT3 had very little effect on TG metabolism in HepG2 cells. Additionally, we demonstrated that the MGAT activity of DGAT1 only makes a minor contribution to TG synthesis in intact HepG2 cells. Our data demonstrated that the MGAT pathway has a role in hepatic lipid metabolism with MGAT2, more so than MGAT3, contributing to TG synthesis and secretion.
Collapse
|
11
|
Feng X, Xiao Y, Guo Q, Peng H, Zhou HY, Wang JP, Xia ZY. Parathyroid hormone alleviates non-alcoholic liver steatosis via activating the hepatic cAMP/PKA/CREB pathway. Front Endocrinol (Lausanne) 2022; 13:899731. [PMID: 36060945 PMCID: PMC9428460 DOI: 10.3389/fendo.2022.899731] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), hallmarked by liver steatosis, is becoming a global concern, but effective and safe drugs for NAFLD are still lacking at present. Parathyroid hormone (PTH), the only FDA-approved anabolic treatment for osteoporosis, is important in calcium-phosphate homeostasis. However, little is known about its potential therapeutic effects on other diseases. Here, we report that intermittent administration of PTH ameliorated non-alcoholic liver steatosis in diet-induced obese (DIO) mice and db/db mice, as well as fasting-induced hepatic steatosis. In vitro, PTH inhibits palmitic acid-induced intracellular lipid accumulation in a parathyroid hormone 1 receptor (PTH1R)-dependent manner. Mechanistically, PTH upregulates the expression of genes involved in lipid β-oxidation and suppresses the expression of genes related to lipid uptake and de novo lipogenesis by activating the cAMP/PKA/CREB pathway. Taken together, our current finding proposes a new therapeutic role of PTH on NAFLD.
Collapse
Affiliation(s)
- Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Jian-Ping Wang
- Department of Endocrinology, The Second Affiliated Hospital of University of South China, Hengyang, China
- *Correspondence: Zhu-Ying Xia, ; Jian-Ping Wang,
| | - Zhu-Ying Xia
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Zhu-Ying Xia, ; Jian-Ping Wang,
| |
Collapse
|
12
|
Moon J, Kim P. Intravital Two-photon Imaging of Dynamic Alteration of Hepatic Lipid Droplets in Fasted and Refed State. J Lipid Atheroscler 2021; 10:313-321. [PMID: 34621702 PMCID: PMC8473963 DOI: 10.12997/jla.2021.10.3.313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
Objective The liver plays a central role in lipid metabolism. During fasting and feeding, the fatty acid trafficking between adipose tissue and liver induces accumulation and dissociation of dynamic hepatic lipid droplets (LDs). Herein, we established an intravital 2-photon imaging technique to longitudinally visualize the dynamic in vivo alteration of hepatic LD deposition during fasting and refeeding in the liver of live mouse. Methods Intravital 2-photon imaging of liver was performed to observe hepatic LD alteration induced by fasting for different periods of time, 12, 24, and 48 hours followed by refeeding. Hepatic LDs were fluorescently labelled in vivo by intravenous injection of Seoul-Flour 44 and visualized by custom-built intravital 2-photon microscope. Results Significant increases of the number and size of hepatic LDs were observed by intravital 2-photon imaging of the liver after 12 hours of fasting. The degree of hepatic LD accumulation continuously increased with fasting up to 48 hours. Remarkably, with refeeding for 24 hours, the hepatic LDs accumulated by fasting were fully dissociated and the LD occupancy in the liver was recovered to the normal state. Conclusion Utilizing intravital 2-photon microscope with in vivo systemic fluorescent labeling of LD in live mice, dynamic alterations of hepatic LDs such as accumulation and dissociation by fasting and refeeding were successfully visualized at a subcellular level in vivo. The established method enabling the in vivo visualization of LDs will be a useful tool to investigate the pathophysiology of various diseases associated with dysregulated lipid metabolism.
Collapse
Affiliation(s)
- Jieun Moon
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
13
|
Freitas-Lima LC, Budu A, Estrela GR, da Silva TA, Arruda AC, de Carvalho Araujo R. Metabolic fasting stress is ameliorated in Kinin B1 receptor-deficient mice. Life Sci 2021; 294:120007. [PMID: 34600938 DOI: 10.1016/j.lfs.2021.120007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
The liver has an essential role in responding to metabolic demands under stress conditions. The organ stores, releases, and recycles metabolism-related substrates. However, it is not clear how the Kallikrein-Kinin System modulates metabolic flexibility shift between energetic sources. AIMS To analyze the hepatic metabolism in kinin B1 receptor deficient mice (B1KO mice) under fasting conditions. MAIN METHODS WT and B1KO male mice were allocated in a calorimetric cage for 7 days and 48 h before the euthanasia, half of the animals of both groups were under fasting conditions. Biochemical parameters, ketone bodies (KB), and gene expression involving the liver energetic metabolism genes were evaluated. KEY FINDINGS Kinin B1 receptor (B1R) modulates the metabolic shift under fasting conditions, reducing the VO2 expenditure. A preference for carbohydrates as an energetic source is suggested, as the B1KO group did not display an increase in KB in the serum. Moreover, the B1KO animals displayed higher serum triglycerides concentration compared to WT fasting mice. Interestingly, the lack of B1R induces the increase expression of enzymes from the glycolysis and lipolysis pathways under the fed. However, under fasting, the enzymatic expression of gluconeogenesis, glyceroneogenesis, and ketogenesis of these pathways does not occur, suggesting an absence of the shift metabolism responsivity, and this condition is modulated by PDK4 under FOXO1 control. SIGNIFICANCE B1R has an important role in the hepatic glucose metabolism, which in turn influences the energetic metabolism, and in long-term outcomes, such as in the decrease in hepatic glycogen stores and in the enhancement of hepatic metabolism.
Collapse
Affiliation(s)
| | - Alexandre Budu
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil.
| | - Gabriel Rufino Estrela
- Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil; Department of Clinical and Experimental Oncology, Discipline of Hematology and Hematotherapy, Federal University of São Paulo, 04037002 São Paulo, Brazil.
| | - Thais Alves da Silva
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil.
| | - Adriano Cleis Arruda
- Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil
| | - Ronaldo de Carvalho Araujo
- Department of Biophysics, Federal University of São Paulo, 04039032 São Paulo, Brazil; Department of Medicine, Discipline of Nephrology, Federal University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
14
|
Towards Understanding the Direct and Indirect Actions of Growth Hormone in Controlling Hepatocyte Carbohydrate and Lipid Metabolism. Cells 2021; 10:cells10102532. [PMID: 34685512 PMCID: PMC8533955 DOI: 10.3390/cells10102532] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 02/06/2023] Open
Abstract
Growth hormone (GH) is critical for achieving normal structural growth. In addition, GH plays an important role in regulating metabolic function. GH acts through its GH receptor (GHR) to modulate the production and function of insulin-like growth factor 1 (IGF1) and insulin. GH, IGF1, and insulin act on multiple tissues to coordinate metabolic control in a context-specific manner. This review will specifically focus on our current understanding of the direct and indirect actions of GH to control liver (hepatocyte) carbohydrate and lipid metabolism in the context of normal fasting (sleep) and feeding (wake) cycles and in response to prolonged nutrient deprivation and excess. Caveats and challenges related to the model systems used and areas that require further investigation towards a clearer understanding of the role GH plays in metabolic health and disease are discussed.
Collapse
|
15
|
Galhuber M, Kupper N, Dohr G, Gauster M, Kwapiszewska G, Olschewski A, Jandl K, Gschwandtner E, Schweiger M, Kratky D, Leitinger G, Prokesch A, Kolb D. Simple method of thawing cryo-stored samples preserves ultrastructural features in electron microscopy. Histochem Cell Biol 2021; 155:593-603. [PMID: 33404705 PMCID: PMC8134286 DOI: 10.1007/s00418-020-01952-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 11/25/2022]
Abstract
Preservation of ultrastructural features in biological samples for electron microscopy (EM) is a challenging task that is routinely accomplished through chemical fixation or high-pressure freezing coupled to automated freeze substitution (AFS) using specialized devices. However, samples from clinical (e.g. "biobanking" of bulk biopsies) and preclinical (e.g. whole mouse tissues) specimens are often not specifically prepared for ultrastructural analyses but simply immersed in liquid nitrogen before long-term cryo-storage. We demonstrate that ultrastructural features of such samples are insufficiently conserved using AFS and developed a simple, rapid, and effective method for thawing that does not require specific instrumentation. This procedure consists of dry ice-cooled pre-trimming of frozen tissue and aldehyde fixation for 3 h at 37 °C followed by standard embedding steps. Herein investigated tissues comprised human term placentae, clinical lung samples, as well as mouse tissues of different composition (brown adipose tissue, white adipose tissue, cardiac muscle, skeletal muscle, liver). For all these tissues, we compared electron micrographs prepared from cryo-stored material with our method to images derived from directly prepared fresh tissues with standard chemical fixation. Our protocol yielded highly conserved ultrastructural features and tissue-specific details, largely matching the quality of fresh tissue samples. Furthermore, morphometric analysis of lipid droplets and mitochondria in livers of fasted mice demonstrated that statistically valid quantifications can be derived from samples prepared with our method. Overall, we provide a simple and effective protocol for accurate ultrastructural and morphometric analyses of cryo-stored bulk tissue samples.
Collapse
Affiliation(s)
- Markus Galhuber
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Nadja Kupper
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Gottfried Dohr
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Martin Gauster
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Physiology, LBI for Lung Vascular Research, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria
| | - Andrea Olschewski
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Physiology, LBI for Lung Vascular Research, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria
| | - Katharina Jandl
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Physiology, LBI for Lung Vascular Research, Medical University of Graz, Stiftingtalstrasse 24, 8010 Graz, Austria
| | - Elisabeth Gschwandtner
- Division of Thoracic Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, Biochemistry II, University of Graz, Heinrichstraße 31/II, 8010 Graz, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Gerd Leitinger
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
- Core Facility Ultrastructure Analysis, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| |
Collapse
|
16
|
Niture S, Lin M, Rios-Colon L, Qi Q, Moore JT, Kumar D. Emerging Roles of Impaired Autophagy in Fatty Liver Disease and Hepatocellular Carcinoma. Int J Hepatol 2021; 2021:6675762. [PMID: 33976943 PMCID: PMC8083829 DOI: 10.1155/2021/6675762] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/16/2021] [Accepted: 04/06/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved catabolic process that eliminates dysfunctional cytosolic biomolecules through vacuole-mediated sequestration and lysosomal degradation. Although the molecular mechanisms that regulate autophagy are not fully understood, recent work indicates that dysfunctional/impaired autophagic functions are associated with the development and progression of nonalcoholic fatty liver disease (NAFLD), alcoholic fatty liver disease (AFLD), and hepatocellular carcinoma (HCC). Autophagy prevents NAFLD and AFLD progression through enhanced lipid catabolism and decreasing hepatic steatosis, which is characterized by the accumulation of triglycerides and increased inflammation. However, as both diseases progress, autophagy can become impaired leading to exacerbation of both pathological conditions and progression into HCC. Due to the significance of impaired autophagy in these diseases, there is increased interest in studying pathways and targets involved in maintaining efficient autophagic functions as potential therapeutic targets. In this review, we summarize how impaired autophagy affects liver function and contributes to NAFLD, AFLD, and HCC progression. We will also explore how recent discoveries could provide novel therapeutic opportunities to effectively treat these diseases.
Collapse
Affiliation(s)
- Suryakant Niture
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Minghui Lin
- The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China 750021
| | - Leslimar Rios-Colon
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Qi Qi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - John T. Moore
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| | - Deepak Kumar
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University Durham, NC 27707, USA
| |
Collapse
|
17
|
Metabolomic biomarkers of polycystic ovary syndrome related-obesity: a review of the literature. REV ROMANA MED LAB 2020. [DOI: 10.2478/rrlm-2020-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Background and objectives: Polycystic ovary syndrome (PCOS) displays a phenotype-dependent cardio-metabolic risk. By performing a systematic search of the literature, we aimed to summarize metabolomic signatures associated with obesity in PCOS women.
Data sources and study eligibility criteria: We conducted a comprehensive search including: Embase, PubMed, and Web of Science until 31st of May 2019. We used the terms: metabolomics and polycystic ovary syndrome. We excluded the following papers: animal studies, studies that included only lean PCOS women, reviews, meta-analyses, results of interventional studies, those that did not apply metabolomic techniques.
Results: The lipid signature in obese women with PCOS showed increased levels of free fatty acids (carnitine, adipic acid, linoleic acid, oleic acid) and lower levels of lysophosphatidylcholines and glycerolphosphocholine compared with non-obese PCOS women. Regarding carbohydrate metabolism, a decrease in citric and lactic acid levels characterized obese PCOS women. Decreased lactic acid in obese PCOS women suggests augmented insulin stimulated glucose muscle use in lean, but not in obese women. Considering amino acid metabolomic markers, valine, glycine, serine, threonine, isoleucine and lysine were higher in obese PCOS women. Patients with visceral obesity presented a diminished uptake of essential amino acids, BCAA, leucine and serine in the skeletal muscle. α-ketoglutarate was significantly higher in obese women with PCOS in comparison with lean women with PCOS, distinguishing these 2 subgroups of PCOS with high ‘predictive accuracy’.
Limitations: Overall, a small number of studies have focused on the impact of obesity on the metabolic fingerprints of PCOS women. There is need for properly controlled, high-quality studies.
Conclusions: There is compelling evidence of significant alterations in carbohydrate, lipid, and amino acid metabolism in women with PCOS and obesity. Metabolomics may identify new metabolic pathways involved in PCOS and improve our understanding of the complex relation between PCOS and obesity in order to personalize PCOS therapy.
Collapse
|
18
|
The food contaminant deoxynivalenol provokes metabolic impairments resulting in non-alcoholic fatty liver (NAFL) in mice. Sci Rep 2020; 10:12072. [PMID: 32694515 PMCID: PMC7374573 DOI: 10.1038/s41598-020-68712-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022] Open
Abstract
The ribotoxin deoxynivalenol (DON) is a trichothecene found on cereals responsible for mycotoxicosis in both humans and farm animals. DON toxicity is characterized by reduced food intake, diminished nutritional efficiency and immunologic effects. The present study was designed to further characterize the alterations in energy metabolism induced by DON intoxication. We demonstrated that acute DON intoxication triggered liver steatosis associated with an altered expression of genes related to lipids oxidation, lipogenesis and lipolysis. This steatosis was concomitant to anorexia, hypoglycemia and a paradoxical transient insulin release. DON treatment resulted also in stimulation of central autonomic network regulating sympathetic outflow and adrenaline and glucocorticoids secretion. Furthermore, an increased expression of genes linked to inflammation and reticulum endoplasmic stress was observed in the liver of DON-treated mice. Finally, we propose that lipids mobilization from adipose tissues (AT) induced by DON intoxication drives hepatic steatosis since (1) genes encoding lipolytic enzymes were up-regulated in AT and (2) plasma concentration of triglycerides (TGs) and non-esterified fatty acids were increased during DON intoxication. Altogether, these data demonstrate that DON induced hormonal and metabolic dysregulations associated with a spectrum of hepatic abnormalities, evocative of a non-alcoholic fatty liver disease.
Collapse
|
19
|
Key CCC, Bishop AC, Wang X, Zhao Q, Chen GY, Quinn MA, Zhu X, Zhang Q, Parks JS. Human GDPD3 overexpression promotes liver steatosis by increasing lysophosphatidic acid production and fatty acid uptake. J Lipid Res 2020; 61:1075-1086. [PMID: 32430316 DOI: 10.1194/jlr.ra120000760] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
The glycerol phosphate pathway produces more than 90% of the liver triacylglycerol (TAG). LysoPA, an intermediate in this pathway, is produced by glycerol-3-phosphate acyltransferase. Glycerophosphodiester phosphodiesterase domain containing 3 (GDPD3), whose gene was recently cloned, contains lysophospholipase D activity, which produces LysoPA from lysophospholipids. Whether human GDPD3 plays a role in hepatic TAG homeostasis is unknown. We hypothesized that human GDPD3 increases LysoPA production and availability in the glycerol phosphate pathway, promoting TAG biosynthesis. To test our hypothesis, we infected C57BL/6J mice with adeno-associated virus encoding a hepatocyte-specific albumin promoter that drives GFP (control) or FLAG-tagged human GDPD3 overexpression and fed the mice chow or a Western diet to induce hepatosteatosis. Hepatic human GDPD3 overexpression induced LysoPA production and increased FA uptake and incorporation into TAG in mouse hepatocytes and livers, ultimately exacerbating Western diet-induced liver steatosis. Our results also showed that individuals with hepatic steatosis have increased GDPD3 mRNA levels compared with individuals without steatosis. Collectively, these findings indicate that upregulation of GDPD3 expression may play a key role in hepatic TAG accumulation and may represent a molecular target for managing hepatic steatosis.
Collapse
Affiliation(s)
- Chia-Chi C Key
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157. mailto:
| | - Andrew C Bishop
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xianfeng Wang
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Qingxia Zhao
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Guan-Yuan Chen
- Department of Chemistry and Center for Translational Biomedical Research, University of North Carolina at Greensboro, Greensboro, NC 27402
| | - Matthew A Quinn
- Section on Comparative Medicine, Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Xuewei Zhu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157
| | - Qibin Zhang
- Department of Chemistry and Center for Translational Biomedical Research, University of North Carolina at Greensboro, Greensboro, NC 27402
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157; Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27157
| |
Collapse
|
20
|
Neuschwander-Tetri BA. Therapeutic Landscape for NAFLD in 2020. Gastroenterology 2020; 158:1984-1998.e3. [PMID: 32061596 DOI: 10.1053/j.gastro.2020.01.051] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022]
Abstract
Lifestyle modifications focused on healthy eating and regular exercise are the primary recommendations for patients with nonalcoholic steatohepatitis (NASH). However, for multiple societal, psychological, physical, genetic, and epigenetic reasons, the ability of people to adopt and sustain such changes is challenging and typically not successful. To end the epidemic of NASH and prevent its complications, including cirrhosis and hepatocellular carcinoma, pharmacological interventions are now being evaluated in clinical trials. Treatments include drugs targeting energy intake, energy disposal, lipotoxic liver injury, and the resulting inflammation and fibrogenesis that lead to cirrhosis. It is likely that patients develop the phenotype of NASH by multiple mechanisms, and thus the optimal treatments of NASH will likely evolve to personalized therapy once we understand the mechanistic underpinnings of NASH in each patient. Reviewed here is the treatment landscape in this rapidly evolving field with an emphasis on drugs in Phase 2 and Phase 3 trials.
Collapse
|
21
|
Dedual MA, Wueest S, Borsigova M, Konrad D. Intermittent fasting improves metabolic flexibility in short-term high-fat diet-fed mice. Am J Physiol Endocrinol Metab 2019; 317:E773-E782. [PMID: 31503513 DOI: 10.1152/ajpendo.00187.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Four days of high-fat diet (HFD) feeding are sufficient to induce glucose intolerance and hepatic steatosis in mice. While prolonged HFD-induced metabolic complications are partly mediated by increased food intake during the light (inactive) phase, such a link has not yet been established in short-term HFD-fed mice. Herein, we hypothesized that a short bout of HFD desynchronizes feeding behavior, thereby contributing to glucose intolerance and hepatic steatosis. To this end, 12-wk-old C57BL/6J littermates were fed a HFD for 4 days either ad libitum or intermittently. Intermittent-fed mice were fasted for 8 h during their inactive phase. Initiation of HFD led to an immediate increase in food intake already during the first light phase. Moreover, glucose tolerance was significantly impaired in ad libitum- but not in intermittent HFD-fed mice, indicating that desynchronized feeding behavior contributes to short-term HFD-induced glucose intolerance. Of note, overall food intake was similar between the groups, as was body weight. However, intermittent HFD-fed mice revealed higher fat depot weights. Phosphorylation of hormone sensitivity lipase and free fatty acid release from isolated adipocytes were significantly elevated, suggesting increased lipolysis in intermittent HFD-fed mice. Moreover, hepatic mRNA expression of lipogenetic enzymes and liver triglyceride levels were significantly increased in intermittent HFD-fed mice. Importantly, food deprivation decreased respiratory exchange ratio promptly in intermittent- but not in ad libitum HFD-fed mice. In conclusion, retaining a normal feeding pattern prevented HFD-induced impairment of metabolic flexibility in short-term HFD-fed mice.
Collapse
Affiliation(s)
- Mara A Dedual
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Marcela Borsigova
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Olivares S, Henkel AS. The role of X-box binding protein 1 in the hepatic response to refeeding in mice. J Lipid Res 2018; 60:353-359. [PMID: 30482806 DOI: 10.1194/jlr.m086413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 11/08/2018] [Indexed: 02/03/2023] Open
Abstract
Refeeding mice after a prolonged fast is a potent stimulus of hepatic lipogenesis, but is also associated with induction of the hepatic unfolded protein response (UPR). The X-box binding protein 1 (Xbp1), a key regulator of the adaptive UPR, transcriptionally activates hepatic lipogenesis genes. We therefore determined whether hepatic Xbp1 mediates the hepatic lipogenic response to refeeding. Mice bearing a hepatocyte-specific deletion of Xbp1 and littermate controls were fasted overnight, followed by refeeding for up to 6 h. Among control mice, refeeding induced hepatic expression of activated Xbp1 and, as expected, induced hepatic expression of genes controlling de novo lipogenesis of fatty acids. Unexpectedly, deletion of hepatic Xbp1 allowed for normal induction of hepatic lipogenesis genes, yet impaired translation of SREBP1c and its targets in response to refeeding. Impaired protein translation was associated with enhanced postprandial activation of the global translational arrest protein, eukaryotic initiation factor 2α, among mice lacking hepatic Xbp1 Deletion of hepatic Xbp1 prevented postprandial induction of genes regulating protein folding and processing and shifted the pattern of postprandial UPR activation to favor proapoptotic signals. We conclude that activation of hepatic Xbp1 in the postprandial states serves the dual roles of restoring postprandial hepatic lipogenesis and proteostasis.
Collapse
Affiliation(s)
- Shantel Olivares
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Anne S Henkel
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL .,Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL
| |
Collapse
|
23
|
Tenopoulou M, Doulias PT, Nakamoto K, Berrios K, Zura G, Li C, Faust M, Yakovishina V, Evans P, Tan L, Bennett MJ, Snyder NW, Quinn WJ, Baur JA, Atochin DN, Huang PL, Ischiropoulos H. Oral nitrite restores age-dependent phenotypes in eNOS-null mice. JCI Insight 2018; 3:122156. [PMID: 30135317 DOI: 10.1172/jci.insight.122156] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023] Open
Abstract
Alterations in the synthesis and bioavailability of NO are central to the pathogenesis of cardiovascular and metabolic disorders. Although endothelial NO synthase-derived (eNOS-derived) NO affects mitochondrial long-chain fatty acid β-oxidation, the pathophysiological significance of this regulation remains unclear. Accordingly, we determined the contributions of eNOS/NO signaling in the adaptive metabolic responses to fasting and in age-induced metabolic dysfunction. Four-month-old eNOS-/- mice are glucose intolerant and exhibit serum dyslipidemia and decreased capacity to oxidize fatty acids. However, during fasting, eNOS-/- mice redirect acetyl-CoA to ketogenesis to elevate circulating levels of β-hydroxybutyrate similar to wild-type mice. Treatment of 4-month-old eNOS-/- mice with nitrite for 10 days corrected the hypertension and serum hyperlipidemia and normalized the rate of fatty acid oxidation. Fourteen-month-old eNOS-/- mice exhibited metabolic derangements, resulting in reduced utilization of fat to generate energy, lower resting metabolic activity, and diminished physical activity. Seven-month administration of nitrite to eNOS-/- mice reversed the age-dependent metabolic derangements and restored physical activity. While the eNOS/NO signaling is not essential for the metabolic adaptation to fasting, it is critical for regulating systemic metabolic homeostasis in aging. The development of age-dependent metabolic disorder is prevented by low-dose replenishment of bioactive NO.
Collapse
Affiliation(s)
- Margarita Tenopoulou
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | | | - Kent Nakamoto
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Kiara Berrios
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Gabriella Zura
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Chenxi Li
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Michael Faust
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Veronika Yakovishina
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Perry Evans
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Lu Tan
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Michael J Bennett
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| | - Nathaniel W Snyder
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, Pennsylvania, USA
| | - William J Quinn
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph A Baur
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dmitriy N Atochin
- Cardiovascular Research Center Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Paul L Huang
- Cardiovascular Research Center Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Harry Ischiropoulos
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Li G, Brocker CN, Yan T, Xie C, Krausz KW, Xiang R, Gonzalez FJ. Metabolic adaptation to intermittent fasting is independent of peroxisome proliferator-activated receptor alpha. Mol Metab 2017; 7:80-89. [PMID: 29146411 PMCID: PMC5784329 DOI: 10.1016/j.molmet.2017.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 10/19/2017] [Accepted: 10/24/2017] [Indexed: 12/15/2022] Open
Abstract
Background Peroxisome proliferator-activated receptor alpha (PPARA) is a major regulator of fatty acid oxidation and severe hepatic steatosis occurs during acute fasting in Ppara-null mice. Thus, PPARA is considered an important mediator of the fasting response; however, its role in other fasting regiments such as every-other-day fasting (EODF) has not been investigated. Methods Mice were pre-conditioned using either a diet containing the potent PPARA agonist Wy-14643 or an EODF regimen prior to acute fasting. Ppara-null mice were used to assess the contribution of PPARA activation during the metabolic response to EODF. Livers were collected for histological, biochemical, qRT-PCR, and Western blot analysis. Results Acute fasting activated PPARA and led to steatosis, whereas EODF protected against fasting-induced hepatic steatosis without affecting PPARA signaling. In contrast, pretreatment with Wy-14,643 did activate PPARA signaling but did not ameliorate acute fasting-induced steatosis and unexpectedly promoted liver injury. Ppara ablation exacerbated acute fasting-induced hypoglycemia, hepatic steatosis, and liver injury in mice, whereas these detrimental effects were absent in response to EODF, which promoted PPARA-independent fatty acid metabolism and normalized serum lipids. Conclusions These findings indicate that PPARA activation prior to acute fasting cannot ameliorate fasting-induced hepatic steatosis, whereas EODF induced metabolic adaptations to protect against fasting-induced steatosis without altering PPARA signaling. Therefore, PPARA activation does not mediate the metabolic adaptation to fasting, at least in preventing acute fasting-induced steatosis. Wy-14,643 activates PPARA but does not alleviate acute fasting-induced steatosis. EODF prevents acute fasting-induced steatosis but does not activate PPARA. EODF protects against fasting-induced steatosis, even in Ppara-null mice. EODF normalizes serum acylcarnitines in Ppara-null mice.
Collapse
Affiliation(s)
- Guolin Li
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Laboratory of Aging Biochemistry, College of Life Sciences, Hunan Normal University, Changsha 410081, China; The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rong Xiang
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha 41001, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
BasuRay S, Smagris E, Cohen JC, Hobbs HH. The PNPLA3 variant associated with fatty liver disease (I148M) accumulates on lipid droplets by evading ubiquitylation. Hepatology 2017; 66:1111-1124. [PMID: 28520213 PMCID: PMC5605398 DOI: 10.1002/hep.29273] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/08/2017] [Accepted: 05/11/2017] [Indexed: 12/14/2022]
Abstract
UNLABELLED A sequence variation (I148M) in patatin-like phospholipase domain-containing protein 3 (PNPLA3) is strongly associated with fatty liver disease, but the underlying mechanism remains obscure. In this study, we used knock-in (KI) mice (Pnpla3148M/M ) to examine the mechanism responsible for accumulation of triglyceride (TG) and PNPLA3 in hepatic lipid droplets (LDs). No differences were found between Pnpla3148M/M and Pnpla3+/+ mice in hepatic TG synthesis, utilization, or secretion. These results are consistent with TG accumulation in the Pnpla3148M/M mice being caused by impaired TG mobilization from LDs. Sucrose feeding, which is required to elicit fatty liver in KI mice, led to a much larger and more persistent increase in PNPLA3 protein in the KI mice than in wild-type (WT) mice. Inhibition of the proteasome (bortezomib), but not macroautophagy (3-methyladenine), markedly increased PNPLA3 levels in WT mice, coincident with the appearance of ubiquitylated forms of the protein. Bortezomib did not increase PNPLA3 levels in Pnpla3148M/M mice, and only trace amounts of ubiquitylated PNPLA3 were seen in these animals. CONCLUSION These results are consistent with the notion that the 148M variant disrupts ubiquitylation and proteasomal degradation of PNPLA3, resulting in accumulation of PNPLA3-148M and impaired mobilization of TG from LDs. (Hepatology 2017;66:1111-1124).
Collapse
Affiliation(s)
- Soumik BasuRay
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasTX,Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX
| | - Eriks Smagris
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasTX,Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX
| | - Jonathan C. Cohen
- Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX
| | - Helen H. Hobbs
- Department of Molecular GeneticsUniversity of Texas Southwestern Medical CenterDallasTX,Department of Internal MedicineUniversity of Texas Southwestern Medical CenterDallasTX,Howard Hughes Medical InstituteUniversity of Texas Southwestern Medical CenterDallasTX
| |
Collapse
|
26
|
Acetylation of Cavin-1 Promotes Lipolysis in White Adipose Tissue. Mol Cell Biol 2017; 37:MCB.00058-17. [PMID: 28559430 DOI: 10.1128/mcb.00058-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 05/19/2017] [Indexed: 01/14/2023] Open
Abstract
White adipose tissue (WAT) serves as a reversible energy storage depot in the form of lipids in response to nutritional status. Cavin-1, an essential component in the biogenesis of caveolae, is a positive regulator of lipolysis in adipocytes. However, molecular mechanisms of cavin-1 in the modulation of lipolysis remain poorly understood. Here, we showed that cavin-1 was acetylated at lysines 291, 293, and 298 (3K), which were under nutritional regulation in WAT. We further identified GCN5 as the acetyltransferase and Sirt1 as the deacetylase of cavin-1. Acetylation-mimetic 3Q mutants of cavin-1 augmented fat mobilization in 3T3-L1 adipocytes and zebrafish. Mechanistically, acetylated cavin-1 preferentially interacted with hormone-sensitive lipase and recruited it to the caveolae, thereby promoting lipolysis. Our findings shed light on the essential role of cavin-1 in regulating lipolysis in an acetylation-dependent manner in WAT.
Collapse
|
27
|
Zhang X, Heckmann BL, Campbell LE, Liu J. G0S2: A small giant controller of lipolysis and adipose-liver fatty acid flux. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28645852 DOI: 10.1016/j.bbalip.2017.06.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The discovery of adipose triglyceride lipase (ATGL) and its coactivator comparative gene identification-58 (CGI-58) provided a major paradigm shift in the understanding of intracellular lipolysis in both adipocytes and nonadipocyte cells. The subsequent discovery of G0/G1 switch gene 2 (G0S2) as a potent endogenous inhibitor of ATGL revealed a unique mechanism governing lipolysis and fatty acid (FA) availability. G0S2 is highly conserved in vertebrates, and exhibits cyclical expression pattern between adipose tissue and liver that is critical to lipid flux and energy homeostasis in these two tissues. Biochemical and cell biological studies have demonstrated that a direct interaction with ATGL mediates G0S2's inhibitory effects on lipolysis and lipid droplet degradation. In this review we examine evidence obtained from recent in vitro and in vivo studies that lends support to the proof-of-principle concept that G0S2 functions as a master regulator of tissue-specific balance of TG storage vs. mobilization, partitioning of metabolic fuels between adipose and liver, and the whole-body adaptive energy response. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Latoya E Campbell
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Jun Liu
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States; Division of Endocrinology, Mayo Clinic, Scottsdale, AZ, United States.
| |
Collapse
|
28
|
Acute Fasting Induces Expression of Acylglycerophosphate Acyltransferase (AGPAT) Enzymes in Murine Liver, Heart, and Brain. Lipids 2017; 52:457-461. [DOI: 10.1007/s11745-017-4251-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 03/28/2017] [Indexed: 10/19/2022]
|
29
|
Heckmann BL, Zhang X, Saarinen AM, Schoiswohl G, Kershaw EE, Zechner R, Liu J. Liver X receptor α mediates hepatic triglyceride accumulation through upregulation of G0/G1 Switch Gene 2 expression. JCI Insight 2017; 2:e88735. [PMID: 28239648 DOI: 10.1172/jci.insight.88735] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Liver X receptors (LXRs) are transcription factors essential for cholesterol homeostasis and lipogenesis. LXRα has been implicated in regulating hepatic triglyceride (TG) accumulation upon both influx of adipose-derived fatty acids (FAs) during fasting and stimulation of de novo FA synthesis by chemical agonism of LXR. However, whether or not a convergent mechanism is employed to drive deposition of FAs from these 2 different sources in TGs is undetermined. Here, we report that the G0/G1 Switch Gene 2 (G0S2), a selective inhibitor of intracellular TG hydrolysis/lipolysis, is a direct target gene of LXRα. Transcriptional activation is conferred by LXRα binding to a direct repeat 4 (DR4) motif in the G0S2 promoter. While LXRα-/- mice exhibited decreased hepatic G0S2 expression, adenoviral expression of G0S2 was sufficient to restore fasting-induced TG storage and glycogen depletion in the liver of these mice. In response to LXR agonist T0901317, G0S2 ablation prevented hepatic steatosis and hypertriglyceridemia without affecting the beneficial effects on HDL. Thus, the LXRα-G0S2 axis plays a distinct role in regulating hepatic TG during both fasting and pharmacological activation of LXR.
Collapse
Affiliation(s)
- Bradlee L Heckmann
- Department of Biochemistry and Molecular Biology.,HEALth Program, Mayo Clinic in Arizona, Scottsdale, Arizona, USA.,Mayo Graduate School, Rochester, Minnesota, USA
| | - Xiaodong Zhang
- Department of Biochemistry and Molecular Biology.,HEALth Program, Mayo Clinic in Arizona, Scottsdale, Arizona, USA
| | - Alicia M Saarinen
- Department of Biochemistry and Molecular Biology.,HEALth Program, Mayo Clinic in Arizona, Scottsdale, Arizona, USA
| | - Gabriele Schoiswohl
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Erin E Kershaw
- Division of Endocrinology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Jun Liu
- Department of Biochemistry and Molecular Biology.,HEALth Program, Mayo Clinic in Arizona, Scottsdale, Arizona, USA.,Division of Endocrinology, Mayo Clinic in Arizona, Scottsdale, Arizona, USA
| |
Collapse
|
30
|
Prokesch A, Graef FA, Madl T, Kahlhofer J, Heidenreich S, Schumann A, Moyschewitz E, Pristoynik P, Blaschitz A, Knauer M, Muenzner M, Bogner-Strauss JG, Dohr G, Schulz TJ, Schupp M. Liver p53 is stabilized upon starvation and required for amino acid catabolism and gluconeogenesis. FASEB J 2017; 31:732-742. [PMID: 27811061 PMCID: PMC5240663 DOI: 10.1096/fj.201600845r] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/24/2016] [Indexed: 12/17/2022]
Abstract
The ability to adapt cellular metabolism to nutrient availability is critical for survival. The liver plays a central role in the adaptation to starvation by switching from glucose-consuming processes and lipid synthesis to providing energy substrates like glucose to the organism. Here we report a previously unrecognized role of the tumor suppressor p53 in the physiologic adaptation to food withdrawal. We found that starvation robustly increases p53 protein in mouse liver. This induction was posttranscriptional and mediated by a hepatocyte-autonomous and AMP-activated protein kinase-dependent mechanism. p53 stabilization was required for the adaptive expression of genes involved in amino acid catabolism. Indeed, acute deletion of p53 in livers of adult mice impaired hepatic glycogen storage and induced steatosis. Upon food withdrawal, p53-deleted mice became hypoglycemic and showed defects in the starvation-associated utilization of hepatic amino acids. In summary, we provide novel evidence for a p53-dependent integration of acute changes of cellular energy status and the metabolic adaptation to starvation. Because of its tumor suppressor function, p53 stabilization by starvation could have implications for both metabolic and oncological diseases of the liver.-Prokesch, A., Graef, F. A., Madl, T., Kahlhofer, J., Heidenreich, S., Schumann, A., Moyschewitz, E., Pristoynik, P., Blaschitz, A., Knauer, M., Muenzner, M., Bogner-Strauss, J. G., Dohr, G., Schulz, T. J., Schupp, M. Liver p53 is stabilized upon starvation and required for amino acid catabolism and gluconeogenesis.
Collapse
Affiliation(s)
- Andreas Prokesch
- Institute of Cell Biology, Histology, and Embryology, Medical University Graz, Graz, Austria;
| | - Franziska A Graef
- Institute of Pharmacology, Center for Cardiovascular Research, Charité University Medicine, Berlin, Germany
| | - Tobias Madl
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Graz, Austria
| | - Jennifer Kahlhofer
- Institute of Cell Biology, Histology, and Embryology, Medical University Graz, Graz, Austria
| | - Steffi Heidenreich
- Institute of Pharmacology, Center for Cardiovascular Research, Charité University Medicine, Berlin, Germany
| | - Anne Schumann
- Institute of Pharmacology, Center for Cardiovascular Research, Charité University Medicine, Berlin, Germany
| | - Elisabeth Moyschewitz
- Institute of Cell Biology, Histology, and Embryology, Medical University Graz, Graz, Austria
| | - Petra Pristoynik
- Institute of Cell Biology, Histology, and Embryology, Medical University Graz, Graz, Austria
| | - Astrid Blaschitz
- Institute of Cell Biology, Histology, and Embryology, Medical University Graz, Graz, Austria
| | - Miriam Knauer
- Institute of Pharmacology, Center for Cardiovascular Research, Charité University Medicine, Berlin, Germany
| | - Matthias Muenzner
- Institute of Pharmacology, Center for Cardiovascular Research, Charité University Medicine, Berlin, Germany
| | | | - Gottfried Dohr
- Institute of Cell Biology, Histology, and Embryology, Medical University Graz, Graz, Austria
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbruecke, Germany; and
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Michael Schupp
- Institute of Pharmacology, Center for Cardiovascular Research, Charité University Medicine, Berlin, Germany;
| |
Collapse
|
31
|
Kwanten WJ, Vandewynckel YP, Martinet W, De Winter BY, Michielsen PP, Van Hoof VO, Driessen A, Timmermans JP, Bedossa P, Van Vlierberghe H, Francque SM. Hepatocellular autophagy modulates the unfolded protein response and fasting-induced steatosis in mice. Am J Physiol Gastrointest Liver Physiol 2016; 311:G599-G609. [PMID: 27514483 PMCID: PMC5142201 DOI: 10.1152/ajpgi.00418.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/05/2016] [Indexed: 01/31/2023]
Abstract
Autophagy and the unfolded protein response (UPR) are key cellular homeostatic mechanisms and are both involved in liver diseases, including nonalcoholic fatty liver disease (NAFLD). Although increasing but conflicting results link these mechanisms to lipid metabolism, their role and potential cross talk herein have been poorly investigated. Therefore, we assessed the effects of hepatocyte-specific autophagy deficiency on liver parenchyma, the UPR, and lipid metabolism. Adult hepatocellular-specific autophagy-deficient mice (Atg7F/FAlb-Cre+) were compared with their autophagy-competent littermates (Atg7+/+Alb-Cre+). Livers were analyzed by electron microscopy, histology, real-time qPCR, and Western blotting. Atg7F/FAlb-Cre+ mice developed hepatomegaly with significant parenchymal injury, as shown by inflammatory infiltrates, hepatocellular apoptosis, pericellular fibrosis, and a pronounced ductular reaction. Surprisingly, the UPR exhibited a pathway-selective pattern upon autophagy deficiency. The activity of the adaptive activating transcription factor 6 (ATF6) pathway was abolished, whereas the proapoptotic protein kinase RNA-like ER kinase pathway was increased compared with Atg7+/+Alb-Cre+ mice. The inositol-requiring enzyme-1α signal was unaltered. Fasting-induced steatosis was absent in Atg7F/FAlb-Cre+ mice. Remarkably, some isolated islands of fat-containing and autophagy-competent cells were observed in these livers. Hepatocellular autophagy is essential for parenchymal integrity in mice. Moreover, in the case of autophagy deficiency, the three different UPR branches are pathway selectively modulated. Attenuation of the ATF6 pathway might explain the observed impairment of fasting-induced steatosis. Finally, autophagy and lipid droplets are directly linked to each other.
Collapse
Affiliation(s)
- Wilhelmus J. Kwanten
- 1Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium;
| | | | - Wim Martinet
- 3Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium;
| | - Benedicte Y. De Winter
- 1Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium;
| | - Peter P. Michielsen
- 1Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium; ,4Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem (Antwerp), Belgium;
| | - Viviane O. Van Hoof
- 5Department of Clinical Chemistry, Antwerp University Hospital, Edegem (Antwerp), Belgium;
| | - Ann Driessen
- 6Department of Pathology, Antwerp University Hospital/University of Antwerp, Edegem (Antwerp), Belgium;
| | | | - Pierre Bedossa
- 8Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Paris University-Denis Diderot, Clichy, France
| | - Hans Van Vlierberghe
- 2Department of Hepatology and Gastroenterology, Ghent University, Ghent, Belgium;
| | - Sven M. Francque
- 1Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, Antwerp, Belgium; ,4Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem (Antwerp), Belgium;
| |
Collapse
|
32
|
Choi JH, Pichiah PBT, Kim MJ, Cha YS. Cheonggukjang, a soybean paste fermented with B. licheniformis-67 prevents weight gain and improves glycemic control in high fat diet induced obese mice. J Clin Biochem Nutr 2016; 59:31-8. [PMID: 27499576 PMCID: PMC4933689 DOI: 10.3164/jcbn.15-30] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/16/2015] [Indexed: 12/17/2022] Open
Abstract
In this study, we investigated the anti-obesity effects of soybean paste—Cheonggukjang, fermented with poly gamma glutamic acid producing Bacillus licheniformis-67 in diet induced obese C57BL/6J mice. Forty male C57BL/6J mice aged 4 weeks were divided into four dietary groups; normal diet control, high fat diet control, high fat diet containing 30% of unfermented soybean and high fat diet containing 30% Cheonggukjang fermented with Bacillus licheniformis-67. After 13 weeks of dietary intervention the mice were sacrificed; serum and tissue samples were examined. Serum and hepatic lipid profile, blood glucose, insulin, leptin level were lower (<0.05) along with the body weight and epididymal fat pad weight in the 30% Cheonggukjang supplemented group compared with the high fat diet control group. The expression level of lipid anabolic gene was significantly decreased; whereas the expression level of lipid catabolic genes were significantly increased in the 30% Cheonggukjang supplemented group compared to the high fat diet control group. Collectively, these results suggested that intake of Cheonggukjang fermented with Bacillus licheniformis-67 significantly prevents obesity related parameters.
Collapse
Affiliation(s)
- Joo-Hee Choi
- Department of Food Science and Human Nutrition, Chonbuk National University, 664-14 Duckjin-dong, Jeonju, Jeonbuk 561-756, Korea
| | - P B Tirupathi Pichiah
- Department of Food Science and Human Nutrition, Chonbuk National University, 664-14 Duckjin-dong, Jeonju, Jeonbuk 561-756, Korea
| | - Min-Jung Kim
- Korea Food Research Institute, 1201-62 Anyangpangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-746, Korea
| | - Youn-Soo Cha
- Department of Food Science and Human Nutrition, Chonbuk National University, 664-14 Duckjin-dong, Jeonju, Jeonbuk 561-756, Korea
| |
Collapse
|
33
|
Smagris E, Gilyard S, BasuRay S, Cohen JC, Hobbs HH. Inactivation of Tm6sf2, a Gene Defective in Fatty Liver Disease, Impairs Lipidation but Not Secretion of Very Low Density Lipoproteins. J Biol Chem 2016; 291:10659-76. [PMID: 27013658 PMCID: PMC4865914 DOI: 10.1074/jbc.m116.719955] [Citation(s) in RCA: 165] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 12/18/2022] Open
Abstract
A missense mutation (E167K) in TM6SF2 (transmembrane 6 superfamily member 2), a polytopic protein of unknown function, is associated with the full spectrum of fatty liver disease. To investigate the role of TM6SF2 in hepatic triglyceride (TG) metabolism, we inactivated the gene in mice. Chronic inactivation of Tm6sf2 in mice is associated with hepatic steatosis, hypocholesterolemia, and transaminitis, thus recapitulating the phenotype observed in humans. No dietary challenge was required to elicit the phenotype. Immunocytochemical and cell fractionation studies revealed that TM6SF2 was present in the endoplasmic reticulum and Golgi complex, whereas the excess neutral lipids in the Tm6sf2(-/-) mice were located in lipid droplets. Plasma VLDL-TG levels were reduced in the KO animals due to a 3-fold decrease in VLDL-TG secretion rate without any associated reduction in hepatic apoB secretion. Both VLDL particle size and plasma cholesterol levels were significantly reduced in KO mice. Despite levels of TM6SF2 protein being 10-fold higher in the small intestine than in the liver, dietary lipid absorption was only modestly reduced in the KO mice. Our data, taken together, reveal that TM6SF2 is required to mobilize neutral lipids for VLDL assembly but is not required for secretion of apoB-containing lipoproteins. Despite TM6SF2 being located in the endoplasmic reticulum and Golgi complex, the lipids that accumulate in its absence reside in lipid droplets.
Collapse
Affiliation(s)
- Eriks Smagris
- From the Departments of Molecular Genetics and Internal Medicine and
| | - Shenise Gilyard
- From the Departments of Molecular Genetics and Internal Medicine and
| | | | | | - Helen H Hobbs
- From the Departments of Molecular Genetics and Internal Medicine and the Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
34
|
Fujikawa Y, Roma LP, Sobotta MC, Rose AJ, Diaz MB, Locatelli G, Breckwoldt MO, Misgeld T, Kerschensteiner M, Herzig S, Müller-Decker K, Dick TP. Mouse redox histology using genetically encoded probes. Sci Signal 2016; 9:rs1. [DOI: 10.1126/scisignal.aad3895] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
35
|
Lantier L, Williams AS, Williams IM, Yang KK, Bracy DP, Goelzer M, James FD, Gius D, Wasserman DH. SIRT3 Is Crucial for Maintaining Skeletal Muscle Insulin Action and Protects Against Severe Insulin Resistance in High-Fat-Fed Mice. Diabetes 2015; 64:3081-92. [PMID: 25948682 PMCID: PMC4542443 DOI: 10.2337/db14-1810] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 04/14/2015] [Indexed: 12/11/2022]
Abstract
Protein hyperacetylation is associated with glucose intolerance and insulin resistance, suggesting that the enzymes regulating the acetylome play a role in this pathological process. Sirtuin 3 (SIRT3), the primary mitochondrial deacetylase, has been linked to energy homeostasis. Thus, it is hypothesized that the dysregulation of the mitochondrial acetylation state, via genetic deletion of SIRT3, will amplify the deleterious effects of a high-fat diet (HFD). Hyperinsulinemic-euglycemic clamp experiments show, for the first time, that mice lacking SIRT3 exhibit increased insulin resistance due to defects in skeletal muscle glucose uptake. Permeabilized muscle fibers from HFD-fed SIRT3 knockout (KO) mice showed that tricarboxylic acid cycle substrate-based respiration is decreased while fatty acid-based respiration is increased, reflecting a fuel switch from glucose to fatty acids. Consistent with reduced muscle glucose uptake, hexokinase II (HKII) binding to the mitochondria is decreased in muscle from HFD-fed SIRT3 KO mice, suggesting decreased HKII activity. These results show that the absence of SIRT3 in HFD-fed mice causes profound impairments in insulin-stimulated muscle glucose uptake, creating an increased reliance on fatty acids. Insulin action was not impaired in the lean SIRT3 KO mice. This suggests that SIRT3 protects against dietary insulin resistance by facilitating glucose disposal and mitochondrial function.
Collapse
Affiliation(s)
- Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN
| | - Ashley S Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Ian M Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Karen K Yang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Deanna P Bracy
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Mickael Goelzer
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Freyja D James
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - David Gius
- Departments of Radiation Oncology and Pharmacology and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN
| |
Collapse
|
36
|
Langhi C, Baldán Á. CIDEC/FSP27 is regulated by peroxisome proliferator-activated receptor alpha and plays a critical role in fasting- and diet-induced hepatosteatosis. Hepatology 2015; 61:1227-38. [PMID: 25418138 PMCID: PMC4376564 DOI: 10.1002/hep.27607] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 11/10/2014] [Indexed: 12/16/2022]
Abstract
UNLABELLED The cell death-inducing DNA fragmentation factor alpha-like effector c (CIDEC; also known in rodents as FSP27 or fat-specific protein 27) is a lipid droplet-associated protein that promotes intracellular triglyceride (TAG) storage. CIDEC/Fsp27 is highly expressed in adipose tissue, but undetectable in normal liver. However, its hepatic expression rises during fasting or under genetic or diet-induced hepatosteatosis in both mice and patients. Herein, we demonstrate that CIDEC/Fsp27 is a direct transcriptional target of the nuclear receptor PPARα (peroxisome proliferator-activated receptor alpha) in both mouse and human hepatocytes, and that preventing Fsp27 induction accelerates PPARα-stimulated fatty acid oxidation. We show that adenoviral-mediated silencing of hepatic Fsp27 abolishes fasting-induced liver steatosis in the absence of changes in plasma lipids. Finally, we report that anti-Fsp27 short hairpin RNA and PPARα agonists synergize to ameliorate hepatosteatosis in mice fed a high fat diet. CONCLUSIONS Together, our data highlight the physiological importance of CIDEC/Fsp27 in TAG homeostasis under both physiological and pathological liver steatosis. Our results also suggest that patients taking fibrates likely have elevated levels of hepatic CIDEC, which may limit the efficient mobilization and catabolism of hepatic TAGs.
Collapse
Affiliation(s)
- Cédric Langhi
- Edward A. Doisy Department of Biochemistry & Molecular Biology, Center for Cardiovascular Research, Saint Louis University, Saint Louis, MO
| | | |
Collapse
|
37
|
Short-term food restriction followed by controlled refeeding promotes gorging behavior, enhances fat deposition, and diminishes insulin sensitivity in mice. J Nutr Biochem 2015; 26:721-8. [PMID: 25913018 DOI: 10.1016/j.jnutbio.2015.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/08/2015] [Accepted: 01/23/2015] [Indexed: 01/08/2023]
Abstract
Rodents are commonly used in food restriction refeeding studies to investigate weight regain. Mice that are rationed food every 24 h may consume all allocated food in a short time (gorge) and therefore undergo a brief well-fed period followed by an extended fasted period until the next day's food allotment. These exaggerated metabolic states are not typical in mice fed ad libitum (nibbling). The aim of the current study was to elucidate the intraday and cumulative metabolic consequences of gorging (induced by food restriction) in mice during controlled refeeding. Accordingly, following a temporary food restriction, mice were fed rations similar to intakes of controls fed ad libitum. Temporary food restriction initiated gorging behavior that persisted during refeeding; consequently, metabolism-related measurements were obtained in the gorging mice during their daily fed and fasted metabolic states. Robust differences in adipose tissue lipogenic and inflammatory gene expression were found in the gorging mice by metabolic state (fed versus fasted). Additionally, despite a reduced cumulative food intake compared to mice fed ad libitum, restriction-induced gorging mice had increased intraabdominal fat accumulation, diminished hepatic and peripheral insulin sensitivity, and a gene expression profile favoring lipid deposition. Our findings highlight the intraday differences in gene expression in gorging mice before and after feeding that confound comparisons with mice fed ad libitum, or nibbling. The present study also provides evidence that weight regain following food restriction is associated with cumulative metabolic and behavioral abnormalities in mice.
Collapse
|
38
|
Moffat C, Bhatia L, Nguyen T, Lynch P, Wang M, Wang D, Ilkayeva OR, Han X, Hirschey MD, Claypool SM, Seifert EL. Acyl-CoA thioesterase-2 facilitates mitochondrial fatty acid oxidation in the liver. J Lipid Res 2014; 55:2458-70. [PMID: 25114170 DOI: 10.1194/jlr.m046961] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Acyl-CoA thioesterase (Acot)2 localizes to the mitochondrial matrix and hydrolyses long-chain fatty acyl-CoA into free FA and CoASH. Acot2 is expressed in highly oxi-dative tissues and is poised to modulate mitochondrial FA oxidation (FAO), yet its biological role is unknown. Using a model of adenoviral Acot2 overexpression in mouse liver (Ad-Acot2), we show that Acot2 increases the utilization of FA substrate during the daytime in ad libitum-fed mice, but the nighttime switch to carbohydrate oxidation is similar to control mice. In further support of elevated FAO in Acot2 liver, daytime serum ketones were higher in Ad-Acot2 mice, and overnight fasting led to minimal hepatic steatosis as compared with control mice. In liver mitochondria from Ad-Acot2 mice, phosphorylating O₂ consumption was higher with lipid substrate, but not with nonlipid substrate. This increase depended on whether FA could be activated on the outer mitochondrial membrane, suggesting that the FA released by Acot2 could be effluxed from mitochondria then taken back up again for oxidation. This circuit would prevent the build-up of inhibitory long-chain fatty acyl-CoA esters. Altogether, our findings indicate that Acot2 can enhance FAO, possibly by mitigating the accumulation of FAO intermediates within the mitochondrial matrix.
Collapse
Affiliation(s)
- Cynthia Moffat
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Lavesh Bhatia
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Teresa Nguyen
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Peter Lynch
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Miao Wang
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | - Dongning Wang
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - Xianlin Han
- Diabetes and Obesity Research Center, Sanford-Burnham Medical Research Institute, Orlando, FL 32827
| | - Matthew D Hirschey
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27710
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Erin L Seifert
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
39
|
Kwanten WJ, Martinet W, Michielsen PP, Francque SM. Role of autophagy in the pathophysiology of nonalcoholic fatty liver disease: A controversial issue. World J Gastroenterol 2014; 20:7325-7338. [PMID: 24966603 PMCID: PMC4064078 DOI: 10.3748/wjg.v20.i23.7325] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 12/24/2013] [Accepted: 01/08/2014] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a mechanism involved in cellular homeostasis under basal and stressed conditions delivering cytoplasmic content to the lysosomes for degradation to macronutrients. The potential role of autophagy in disease is increasingly recognised and investigated in the last decade. Nowadays it is commonly accepted that autophagy plays a role in the hepatic lipid metabolism. Hence, dysfunction of autophagy may be an underlying cause of non-alcoholic fatty liver disease. However, controversy of the exact role of autophagy in the lipid metabolism exists: some publications report a lipolytic function of autophagy, whereas others claim a lipogenic function. This review aims to give an update of the present knowledge on autophagy in the hepatic lipid metabolism, hepatic insulin resistance, steatohepatitis and hepatic fibrogenesis.
Collapse
|
40
|
Yu ZR, Ning Y, Yu H, Tang NJ. A HPLC-Q-TOF-MS-based urinary metabolomic approach to identification of potential biomarkers of metabolic syndrome. ACTA ACUST UNITED AC 2014; 34:276-283. [DOI: 10.1007/s11596-014-1271-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/01/2014] [Indexed: 01/03/2023]
|
41
|
Zhang X, Xie X, Heckmann BL, Saarinen AM, Czyzyk TA, Liu J. Targeted disruption of G0/G1 switch gene 2 enhances adipose lipolysis, alters hepatic energy balance, and alleviates high-fat diet-induced liver steatosis. Diabetes 2014; 63:934-46. [PMID: 24194501 PMCID: PMC3931401 DOI: 10.2337/db13-1422] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent biochemical and cell-based studies identified G0/G1 switch gene 2 (G0S2) as an inhibitor of adipose triglyceride lipase (ATGL), a key mediator of intracellular triacylglycerol (TG) mobilization. Here, we show that upon fasting, G0S2 protein expression exhibits an increase in liver and a decrease in adipose tissue. Global knockout of G0S2 in mice enhanced adipose lipolysis and attenuated gain of body weight and adiposity. More strikingly, G0S2 knockout mice displayed a drastic decrease in hepatic TG content and were resistant to high-fat diet (HFD)-induced liver steatosis, both of which were reproduced by liver-specific G0S2 knockdown. Mice with hepatic G0S2 knockdown also showed increased ketogenesis, accelerated gluconeogenesis, and decelerated glycogenolysis. Conversely, overexpression of G0S2 inhibited fatty acid oxidation in mouse primary hepatocytes and caused sustained steatosis in liver accompanied by deficient TG clearance during the fasting-refeeding transition. In response to HFD, there was a profound increase in hepatic G0S2 expression in the fed state. Global and hepatic ablation of G0S2 both led to improved insulin sensitivity in HFD-fed mice. Our findings implicate a physiological role for G0S2 in the control of adaptive energy response to fasting and as a contributor to obesity-associated liver steatosis.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ
- HEAL Program, Mayo Clinic in Arizona, Scottsdale, AZ
| | - Xitao Xie
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ
- HEAL Program, Mayo Clinic in Arizona, Scottsdale, AZ
| | - Bradlee L. Heckmann
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ
- HEAL Program, Mayo Clinic in Arizona, Scottsdale, AZ
- Mayo Graduate School, Rochester, MN
| | - Alicia M. Saarinen
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ
- HEAL Program, Mayo Clinic in Arizona, Scottsdale, AZ
| | - Traci A. Czyzyk
- HEAL Program, Mayo Clinic in Arizona, Scottsdale, AZ
- Department of Physiology and Biomedical Engineering, Mayo Clinic in Arizona, Scottsdale, AZ
| | - Jun Liu
- Department of Biochemistry and Molecular Biology, Mayo Clinic in Arizona, Scottsdale, AZ
- HEAL Program, Mayo Clinic in Arizona, Scottsdale, AZ
- Corresponding author: Jun Liu,
| |
Collapse
|
42
|
Xu J, Donepudi AC, Moscovitz JE, Slitt AL. Keap1-knockdown decreases fasting-induced fatty liver via altered lipid metabolism and decreased fatty acid mobilization from adipose tissue. PLoS One 2013; 8:e79841. [PMID: 24224011 PMCID: PMC3817107 DOI: 10.1371/journal.pone.0079841] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/25/2013] [Indexed: 01/10/2023] Open
Abstract
AIMS The purpose of this study was to determine whether Nrf2 activation, via Keap1-knockdown (Keap1-KD), regulates lipid metabolism and mobilization induced by food deprivation (e.g. fasting). METHODS AND RESULTS Male C57BL/6 (WT) and Keap1-KD mice were either fed ad libitum or food deprived for 24 hours. After fasting, WT mice exhibited a marked increase in hepatic lipid accumulation, but Keap1-KD mice had an attenuated increase of lipid accumulation, along with reduced expression of lipogenic genes (acetyl-coA carboxylase, stearoyl-CoA desaturase-1, and fatty acid synthase) and reduced expression of genes related to fatty acid transport, such as fatty acid translocase/CD36 (CD36) and Fatty acid transport protein (FATP) 2, which may attribute to the reduced induction of Peroxisome proliferator-activated receptor (Ppar) α signaling in the liver. Additionally, enhanced Nrf2 activity by Keap1-KD increased AMP-activated protein kinase (AMPK) phosphorylation in liver. In white adipose tissue, enhanced Nrf2 activity did not change the lipolysis rate by fasting, but reduced expression of fatty acid transporters--CD36 and FATP1, via a PPARα-dependent mechanism, which impaired fatty acid transport from white adipose tissue to periphery circulation system, and resulted in increased white adipose tissue fatty acid content. Moreover, enhanced Nrf2 activity increased glucose tolerance and Akt phosphorylation levels upon insulin administration, suggesting Nrf2 signaling pathway plays a key role in regulating insulin signaling and enhanced insulin sensitivity in skeletal muscle. CONCLUSION Enhanced Nrf2 activity via Keap1-KD decreased fasting-induced steatosis, pointing to an important function of Nrf2 on lipid metabolism under the condition of nutrient deprivation.
Collapse
Affiliation(s)
- Jialin Xu
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, P. R. China
| | - Ajay C. Donepudi
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Jamie E. Moscovitz
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Angela L. Slitt
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
43
|
Dokas J, Chadt A, Nolden T, Himmelbauer H, Zierath JR, Joost HG, Al-Hasani H. Conventional knockout of Tbc1d1 in mice impairs insulin- and AICAR-stimulated glucose uptake in skeletal muscle. Endocrinology 2013; 154:3502-14. [PMID: 23892475 DOI: 10.1210/en.2012-2147] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the obesity-resistant SJL mouse strain, we previously identified a naturally occurring loss-of-function mutation in the gene for Tbc1d1. Characterization of recombinant inbred mice that carried the Tbc1d1(SJL) allele on a C57BL/6J background indicated that loss of TBC1D1 protects from obesity, presumably by increasing the use of fat as energy source. To provide direct functional evidence for an involvement of TBC1D1 in energy substrate metabolism, we generated and characterized conventional Tbc1d1 knockout mice. TBC1D1-deficient mice showed moderately reduced body weight, decreased respiratory quotient, and an elevated resting metabolic rate. Ex vivo analysis of intact isolated skeletal muscle revealed a severe impairment in insulin- and AICAR-stimulated glucose uptake in glycolytic extensor digitorum longus muscle and a substantially increased rate of fatty acid oxidation in oxidative soleus muscle. Our results provide direct evidence that TBC1D1 plays a major role in glucose and lipid utilization, and energy substrate preference in skeletal muscle.
Collapse
Affiliation(s)
- Janine Dokas
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Leibniz Center for Diabetes Research at the Heinrich-Heine-University Düsseldorf, Auf'm Hennekamp 65, 40225 Düsseldorf, Germany.
| | | | | | | | | | | | | |
Collapse
|
44
|
Li JZ, Huang Y, Karaman R, Ivanova PT, Brown HA, Roddy T, Castro-Perez J, Cohen JC, Hobbs HH. Chronic overexpression of PNPLA3I148M in mouse liver causes hepatic steatosis. J Clin Invest 2013; 122:4130-44. [PMID: 23023705 DOI: 10.1172/jci65179] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 08/09/2012] [Indexed: 12/12/2022] Open
Abstract
A genetic variant in PNPLA3 (PNPLA3(I148M)), a triacylglycerol (TAG) hydrolase, is a major risk factor for nonalcoholic fatty liver disease (NAFLD); however, the mechanism underlying this association is not known. To develop an animal model of PNPLA3-induced fatty liver disease, we generated transgenic mice that overexpress similar amounts of wild-type PNPLA3 (PNPLA3(WT)) or mutant PNPLA3 (PNPLA3(I148M)) either in liver or adipose tissue. Overexpression of the transgenes in adipose tissue did not affect liver fat content. Expression of PNPLA3(I148M), but not PNPLA3(WT), in liver recapitulated the fatty liver phenotype as well as other metabolic features associated with this allele in humans. Metabolic studies provided evidence for 3 distinct alterations in hepatic TAG metabolism in PNPLA3(I148M) transgenic mice: increased formation of fatty acids and TAG, impaired hydrolysis of TAG, and relative depletion of TAG long-chain polyunsaturated fatty acids. These findings suggest that PNPLA3 plays a role in remodeling TAG in lipid droplets, as they accumulate in response to food intake, and that the increase in hepatic TAG levels associated with the I148M substitution results from multiple changes in hepatic TAG metabolism. The development of an animal model that recapitulates the metabolic phenotype of the allele in humans provides a new platform in which to elucidate the role of PNLPA3(I148M) in NAFLD.
Collapse
Affiliation(s)
- John Zhong Li
- Department of Molecular Genetics, Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046, USA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Heckmann BL, Zhang X, Xie X, Liu J. The G0/G1 switch gene 2 (G0S2): regulating metabolism and beyond. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:276-81. [PMID: 23032787 DOI: 10.1016/j.bbalip.2012.09.016] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 09/24/2012] [Accepted: 09/26/2012] [Indexed: 02/06/2023]
Abstract
The G0/G1 switch gene 2 (G0S2) was originally identified in blood mononuclear cells following induced cell cycle progression. Translation of G0S2 results in a small basic protein of 103 amino acids in size. It was initially believed that G0S2 mediates re-entry of cells from the G0 to G1 phase of the cell cycle. Recent studies have begun to reveal the functional aspects of G0S2 and its protein product in various cellular settings. To date the best-known function of G0S2 is its direct inhibitory capacity on the rate-limiting lipolytic enzyme adipose triglyceride lipase (ATGL). Other studies have illustrated key features of G0S2 including sub-cellular localization, expression profiles and regulation, and possible functions in cellular proliferation and differentiation. In this review we present the current knowledge base regarding all facets of G0S2, and pose a variety of questions and hypotheses pertaining to future research directions.
Collapse
Affiliation(s)
- Bradlee L Heckmann
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Scottsdale, Arizona 85259, USA
| | | | | | | |
Collapse
|
46
|
Softic S, Kirby M, Berger NG, Shroyer NF, Woods SC, Kohli R. Insulin concentration modulates hepatic lipid accumulation in mice in part via transcriptional regulation of fatty acid transport proteins. PLoS One 2012; 7:e38952. [PMID: 22745692 PMCID: PMC3380053 DOI: 10.1371/journal.pone.0038952] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 05/16/2012] [Indexed: 12/18/2022] Open
Abstract
Background Fatty liver disease (FLD) is commonly associated with insulin resistance and obesity, but interestingly it is also observed at low insulin states, such as prolonged fasting. Thus, we asked whether insulin is an independent modulator of hepatic lipid accumulation. Methods/Principal Findings In mice we induced, hypo- and hyperinsulinemia associated FLD by diet induced obesity and streptozotocin treatment, respectively. The mechanism of free fatty acid induced steatosis was studied in cell culture with mouse liver cells under different insulin concentrations, pharmacological phosphoinositol-3-kinase (PI3K) inhibition and siRNA targeted gene knock-down. We found with in vivo and in vitro models that lipid storage is increased, as expected, in both hypo- and hyperinsulinemic states, and that it is mediated by signaling through either insulin receptor substrate (IRS) 1 or 2. As previously reported, IRS-1 was up-regulated at high insulin concentrations, while IRS-2 was increased at low levels of insulin concentration. Relative increase in either of these insulin substrates, was associated with an increase in liver-specific fatty acid transport proteins (FATP) 2&5, and increased lipid storage. Furthermore, utilizing pharmacological PI3K inhibition we found that the IRS-PI3K pathway was necessary for lipogenesis, while FATP responses were mediated via IRS signaling. Data from additional siRNA experiments showed that knock-down of IRSs impacted FATP levels. Conclusions/Significance States of perturbed insulin signaling (low-insulin or high-insulin) both lead to increased hepatic lipid storage via FATP and IRS signaling. These novel findings offer a common mechanism of FLD pathogenesis in states of both inadequate (prolonged fasting) and ineffective (obesity) insulin signaling.
Collapse
Affiliation(s)
- Samir Softic
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, Riley Hospital for Children, Indiana University, Indianapolis, Indiana, United States of America
| | - Michelle Kirby
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Nicholas G. Berger
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Noah F. Shroyer
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Stephen C. Woods
- Metabolic Diseases Institute, Obesity Research Center, Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rohit Kohli
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
47
|
Asterholm IW, McDonald J, Blanchard PG, Sinha M, Xiao Q, Mistry J, Rutkowski JM, Deshaies Y, Brekken RA, Scherer PE. Lack of "immunological fitness" during fasting in metabolically challenged animals. J Lipid Res 2012; 53:1254-67. [PMID: 22504909 DOI: 10.1194/jlr.m021725] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Subclinical inflammation is frequently associated with obesity. Here, we aim to better define the acute inflammatory response during fasting. To do so, we analyzed representatives of immune-related proteins in circulation and in tissues as potential markers for adipose tissue inflammation and modulation of the immune system. Lipopolysaccharide treatment or high-fat diet led to an increase in circulating serum amyloid (SAA) and α1-acid glycoprotein (AGP), whereas adipsin levels were reduced. Mouse models that are protected against diet-induced challenges, such as adiponectin-overexpressing animals or mice treated with PPARγ agonists, displayed lower SAA levels and higher adip-sin levels. An oral lipid gavage, as well as prolonged fasting, increased circulating SAA concurrent with the elevation of free FA levels. Moreover, prolonged fasting was associated with an increased number of Mac2-positive crown-like structures, an increased capillary permeability, and an increase in several M2-type macrophage markers in adipose tissue. This fasting-induced increase in SAA and M2-type macrophage markers was impaired in metabolically challenged animals. These data suggest that metabolic inflexibility is associated with a lack of "immunological fitness."
Collapse
Affiliation(s)
- Ingrid Wernstedt Asterholm
- Touchstone Diabetes Center, Departments of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hugo SE, Cruz-Garcia L, Karanth S, Anderson RM, Stainier DYR, Schlegel A. A monocarboxylate transporter required for hepatocyte secretion of ketone bodies during fasting. Genes Dev 2012; 26:282-93. [PMID: 22302940 DOI: 10.1101/gad.180968.111] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
To find new genes that influence liver lipid mass, we performed a genetic screen for zebrafish mutants with hepatic steatosis, a pathological accumulation of fat. The red moon (rmn) mutant develops hepatic steatosis as maternally deposited yolk is depleted. Conversely, hepatic steatosis is suppressed in rmn mutants by adequate nutrition. Adult rmn mutants show increased liver neutral lipids and induction of hepatic lipid biosynthetic genes when fasted. Positional cloning of the rmn locus reveals a loss-of-function mutation in slc16a6a (solute carrier family 16a, member 6a), a gene that we show encodes a transporter of the major ketone body β-hydroxybutyrate. Restoring wild-type zebrafish slc16a6a expression or introducing human SLC16A6 in rmn mutant livers rescues the mutant phenotype. Radiotracer analysis confirms that loss of Slc16a6a function causes diversion of liver-trapped ketogenic precursors into triacylglycerol. Underscoring the importance of Slc16a6a to normal fasting physiology, previously fed rmn mutants are more sensitive to death by starvation than are wild-type larvae. Our unbiased, forward genetic approach has found a heretofore unrecognized critical step in fasting energy metabolism: hepatic ketone body transport. Since β-hydroxybutyrate is both a major fuel and a signaling molecule in fasting, the discovery of this transporter provides a new direction for modulating circulating levels of ketone bodies in metabolic diseases.
Collapse
Affiliation(s)
- Sarah E Hugo
- University of Utah Molecular Medicine (U2M2) Program, Division of Endocrinology, Metabolism, and Diabetes, University of Utah, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | |
Collapse
|
49
|
Browning JD, Baxter J, Satapati S, Burgess SC. The effect of short-term fasting on liver and skeletal muscle lipid, glucose, and energy metabolism in healthy women and men. J Lipid Res 2012; 53:577-586. [PMID: 22140269 PMCID: PMC3276482 DOI: 10.1194/jlr.p020867] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 11/22/2011] [Indexed: 01/23/2023] Open
Abstract
Fasting promotes triglyceride (TG) accumulation in lean tissues of some animals, but the effect in humans is unknown. Additionally, fasting lipolysis is sexually dimorphic in humans, suggesting that lean tissue TG accumulation and metabolism may differ between women and men. This study investigated lean tissue TG content and metabolism in women and men during extended fasting. Liver and muscle TG content were measured by magnetic resonance spectroscopy during a 48-h fast in healthy men and women. Whole-body and hepatic carbohydrate, lipid, and energy metabolism were also evaluated using biochemical, calorimetric, and stable isotope tracer techniques. As expected, postabsorptive plasma fatty acids (FAs) were higher in women than in men but increased more rapidly in men with the onset of early starvation. Concurrently, sexual dimorphism was apparent in lean tissue TG accumulation during the fast, occurring in livers of men but in muscles of women. Despite differences in lean tissue TG distribution, men and women had identical fasting responses in whole-body and hepatic glucose and oxidative metabolism. In conclusion, TG accumulated in livers of men but in muscles of women during extended fasting. This sexual dimorphism was related to differential fasting plasma FA concentrations but not to whole body or hepatic utilization of this substrate.
Collapse
Affiliation(s)
- Jeffrey D Browning
- Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX; Department of Internal Medicine, The Advanced Imaging Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX.
| | - Jeannie Baxter
- Department of Internal Medicine, The Advanced Imaging Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX
| | - Santhosh Satapati
- Department of Internal Medicine, The Advanced Imaging Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX
| | - Shawn C Burgess
- Department of Internal Medicine, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX; Department of Internal Medicine, The Advanced Imaging Research Center, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX; Department of Pharmacology, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX.
| |
Collapse
|
50
|
Xu J, Kulkarni SR, Li L, Slitt AL. UDP-glucuronosyltransferase expression in mouse liver is increased in obesity- and fasting-induced steatosis. Drug Metab Dispos 2012; 40:259-66. [PMID: 22031624 PMCID: PMC3263941 DOI: 10.1124/dmd.111.039925] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 10/26/2011] [Indexed: 12/21/2022] Open
Abstract
UDP-glucuronosyltransferases (Ugt) catalyze phase II conjugation reactions with glucuronic acid, which enhances chemical polarity and the elimination from the body. Few studies have addressed whether Ugt expression and activity are affected by liver disease, such as steatosis. The purpose of this study was to determine whether steatosis induced by obesity or fasting could affect liver Ugt mRNA expression and activity. Male C57BL/6J and Lep(ob/ob) (ob/ob) mice were fed ad libitum or food was withheld for 24 h. In steatotic livers of ob/ob mice, Ugt1a1, -1a6, -1a9, -2a3, -3a1, and -3a2 mRNA expression increased. Fasting, which also induced steatosis, increased hepatic Ugt1a1, -1a6, -1a7, -1a9, -2b1, -2b5, -2a3, -3a1, and -3a2 mRNA expression in mouse liver. Likewise, acetaminophen glucuronidation increased by 47% in hepatic microsomes from ob/ob mice compared with that in C57BL/6J mice, but not after fasting. In both steatosis models, Ugt induction was accompanied by increased aryl hydrocarbon receptor, constitutive androstane receptor (CAR), peroxisome proliferator-activated receptor (PPAR)-α, pregnane X receptor, nuclear factor (erythroid-derived 2)-like 2 (Nrf2), and peroxisome proliferator-activated receptor-γ coactivator-1α mRNA expression. In addition, fasting increased CAR, PPAR, and Nrf2 binding activity. The work points to hepatic triglyceride concentrations corresponding with nuclear receptor and Ugt expression. The findings indicate that steatosis significantly alters hepatic Ugt expression and activity, which could have a significant impact on determining circulating hormone levels, drug efficacy, and environmental chemical clearance.
Collapse
Affiliation(s)
- Jialin Xu
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, 41 Lower College Rd., Kingston, RI 02881, USA
| | | | | | | |
Collapse
|