51
|
Feraco A, Gorini S, Armani A, Camajani E, Rizzo M, Caprio M. Exploring the Role of Skeletal Muscle in Insulin Resistance: Lessons from Cultured Cells to Animal Models. Int J Mol Sci 2021; 22:ijms22179327. [PMID: 34502235 PMCID: PMC8430804 DOI: 10.3390/ijms22179327] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle is essential to maintain vital functions such as movement, breathing, and thermogenesis, and it is now recognized as an endocrine organ. Muscles release factors named myokines, which can regulate several physiological processes. Moreover, skeletal muscle is particularly important in maintaining body homeostasis, since it is responsible for more than 75% of all insulin-mediated glucose disposal. Alterations of skeletal muscle differentiation and function, with subsequent dysfunctional expression and secretion of myokines, play a key role in the pathogenesis of obesity, type 2 diabetes, and other metabolic diseases, finally leading to cardiometabolic complications. Hence, a deeper understanding of the molecular mechanisms regulating skeletal muscle function related to energy metabolism is critical for novel strategies to treat and prevent insulin resistance and its cardiometabolic complications. This review will be focused on both cellular and animal models currently available for exploring skeletal muscle metabolism and endocrine function.
Collapse
Affiliation(s)
- Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- PhD Programme in Endocrinological Sciences, Department of Experimental Medicine, University of Rome “La Sapienza”, 00161 Rome, Italy
| | - Manfredi Rizzo
- Promise Department, School of Medicine, University of Palermo, 90127 Palermo, Italy;
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, 00166 Rome, Italy; (A.F.); (S.G.); (A.A.)
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
- Correspondence: ; Tel.: +39-065-225-3419
| |
Collapse
|
52
|
Zhao Y, Albrecht E, Li Z, Schregel J, Sciascia QL, Metges CC, Maak S. Distinct Roles of Perilipins in the Intramuscular Deposition of Lipids in Glutamine-Supplemented, Low-, and Normal-Birth-Weight Piglets. Front Vet Sci 2021; 8:633898. [PMID: 34235195 PMCID: PMC8257002 DOI: 10.3389/fvets.2021.633898] [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: 11/26/2020] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Piglets with low birth weight (LBW) usually have reduced muscle mass and increased lipid deposition compared with their normal-birth-weight (NBW) littermates. Supplementation of piglets with amino acids during the first days of life may improve muscle growth and simultaneously alter the intramuscular lipid deposition. The aim of the current study was to investigate the influence of glutamine (Gln) supplementation during the early suckling period on lipid deposition in the longissimus muscle (MLD) and the role of different perilipin (PLIN) family members in this process. Four groups were generated consisting of 72 male LBW piglets and 72 NBW littermates. Piglets were supplemented with either 1 g Gln/kg body weight or an isonitrogenous amount of alanine (Ala) between days post natum (dpn) 1 and 12. Twelve piglets per group were slaughtered at 5, 12, and 26 dpn, and muscle tissue was collected. Perilipins were localized by immunohistochemistry in muscle sections. The mRNA and protein abundances of PLIN family members and related lipases were quantified by quantitative RT-PCR (qPCR) and western blots, respectively. While PLIN1 was localized around lipid droplets in mature and developing adipocytes, PLIN2 was localized at intramyocellular lipid droplets, PLIN3 and 4 at cell membranes of muscle fibers and adipocytes, and PLIN5 in the cytoplasm of undefined cells. The western blot results indicated higher protein abundances of PLIN2, 3, 4, and 5 in LBW piglets (p < 0.05) at 5 dpn compared with their NBW littermates independent of supplementation, while not directly reflecting the mRNA expression levels. The mRNA abundance of PLIN2 was lower while PLIN4 was higher in piglets at 26 dpn in comparison with piglets at 5 dpn (p < 0.01). Relative mRNA expression of LPL and CGI-58 was lowest in piglets at 5 dpn (p < 0.001). However, ATGL mRNA was not influenced by birth weight or supplementation, but the Spearman correlation coefficient analysis revealed close correlations with PLIN2, 4, and 5 mRNA at 5 and 26 dpn (r > 0.5, p < 0.001). The results indicated the importance of birth weight and age for intramuscular lipid deposition and different roles of PLIN family members in this process, but no clear modulating effect of Gln supplementation.
Collapse
Affiliation(s)
- Yaolu Zhao
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Elke Albrecht
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Zeyang Li
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Johannes Schregel
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Quentin L Sciascia
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Cornelia C Metges
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Steffen Maak
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
53
|
Kashiwada M, Nakaishi S, Usuda A, Miyahara Y, Katsumoto K, Katsura K, Terakado I, Jindo M, Nakajima S, Ogawa S, Sugiyama K, Ochiai W. Analysis of anti-obesity and anti-diabetic effects of acacia bark-derived proanthocyanidins in type 2 diabetes model KKAy mice. J Nat Med 2021; 75:893-906. [PMID: 34120298 DOI: 10.1007/s11418-021-01537-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/01/2021] [Indexed: 10/21/2022]
Abstract
The acacia bark extract derived from Acacia mearnsii De Wild is rich in proanthocyanidins, whose constituent units are robinetinidol, fisetinidol, catechin, and gallocatechin. In this study, we examined the effect of proanthocyanidins on obesity and diabetes using KKAy mice, a type 2 diabetes model. KKAy mice were fed either a low-fat diet, a high-fat diet, or a high-fat diet mixed with an acacia bark extract, a proanthocyanidins fraction, and other fraction for 7 weeks. Monitoring the changes in the body weight revealed that acacia bark extract and proanthocyanidins fraction could prevent excessive weight gain resulting from a high-fat diet. In addition, increases in the fasting blood glucose level due to high-fat diet intake were found to be suppressed by acacia bark extract and proanthocyanidins fraction. Furthermore, proanthocyanidins derived from acacia bark were found to increase the expression of adiponectin in white adipose tissue, which enhances the action of insulin. In addition, acacia bark-derived proanthocyanidins suppressed gluconeogenesis and fatty acid synthesis in the liver, as well as suppressing the decrease in energy production under pathological conditions in skeletal muscle. In addition, acacia bark-derived proanthocyanidins showed AMPK activation and DPP-4 inhibitory action. Therefore, it was suggested that acacia bark-derived proanthocyanidins lowered fasting blood glucose levels through the above mechanism. These results suggest that proanthocyanidins derived from acacia bark are the active ingredients of the anti-obesity and anti-diabetic effects of acacia bark extract.
Collapse
Affiliation(s)
- Mayumi Kashiwada
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Saho Nakaishi
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Ayumi Usuda
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Yumi Miyahara
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Kenta Katsumoto
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Kyoko Katsura
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Izumi Terakado
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Makiko Jindo
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Shihori Nakajima
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Sosuke Ogawa
- Acacia-No-Ki Co., Ltd., 4291-1, Miyauchi, Hatsukaichi-shi, Hiroshima, 738-0034, Japan
| | - Kiyoshi Sugiyama
- Department of Functional Molecule Kinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Wataru Ochiai
- Department of Pharmacokinetics, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo, 142-8501, Japan.
| |
Collapse
|
54
|
Chen K, Wei X, Pariyani R, Kortesniemi M, Zhang Y, Yang B. 1H NMR Metabolomics and Full-Length RNA-Seq Reveal Effects of Acylated and Nonacylated Anthocyanins on Hepatic Metabolites and Gene Expression in Zucker Diabetic Fatty Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:4423-4437. [PMID: 33835816 PMCID: PMC8154569 DOI: 10.1021/acs.jafc.1c00130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 06/01/2023]
Abstract
Anthocyanins have been reported to possess antidiabetic effects. Recent studies indicate acylated anthocyanins have better stability and antioxidative activity compared to their nonacylated counterparts. This study compared the effects of nonacylated and acylated anthocyanins on hepatic gene expression and metabolic profile in diabetic rats, using full-length transcriptomics and 1H NMR metabolomics. Zucker diabetic fatty (ZDF) rats were fed with nonacylated anthocyanin extract from bilberries (NAAB) or acylated anthocyanin extract from purple potatoes (AAPP) at daily doses of 25 and 50 mg/kg body weight for 8 weeks. Both anthocyanin extracts restored the levels of multiple metabolites (glucose, lactate, alanine, and pyruvate) and expression of genes (G6pac, Pck1, Pklr, and Gck) involved in glycolysis and gluconeogenesis. AAPP decreased the hepatic glutamine level. NAAB regulated the expression of Mgat4a, Gstm6, and Lpl, whereas AAPP modified the expression of Mgat4a, Jun, Fos, and Egr1. This study indicated different effects of AAPP and NAAB on the hepatic transcriptomic and metabolic profiles of diabetic rats.
Collapse
Affiliation(s)
- Kang Chen
- Food
Chemistry and Food Development,
Department of Life Technologies, University
of Turku, FI-20014 Turun yliopisto, Finland
| | - Xuetao Wei
- Beijing
Key Laboratory of Toxicological Research and Risk Assessment for Food
Safety, Department of Toxicology, School of Public Health, Beijing University, Beijing 100191, China
| | - Raghunath Pariyani
- Food
Chemistry and Food Development,
Department of Life Technologies, University
of Turku, FI-20014 Turun yliopisto, Finland
| | - Maaria Kortesniemi
- Food
Chemistry and Food Development,
Department of Life Technologies, University
of Turku, FI-20014 Turun yliopisto, Finland
| | - Yumei Zhang
- Department
of Nutrition and Food Hygiene, School of Public Health, Beijing University, Beijing 100191, China
| | - Baoru Yang
- Food
Chemistry and Food Development,
Department of Life Technologies, University
of Turku, FI-20014 Turun yliopisto, Finland
| |
Collapse
|
55
|
Presby DM, Rudolph MC, Sherk VD, Jackman MR, Foright RM, Jones KL, Houck JA, Johnson GC, Higgins JA, Neufer PD, Eckel RH, MacLean PS. Lipoprotein Lipase Overexpression in Skeletal Muscle Attenuates Weight Regain by Potentiating Energy Expenditure. Diabetes 2021; 70:867-877. [PMID: 33536195 PMCID: PMC7980196 DOI: 10.2337/db20-0763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 01/27/2021] [Indexed: 11/13/2022]
Abstract
Moderate weight loss improves numerous risk factors for cardiometabolic disease; however, long-term weight loss maintenance (WLM) is often thwarted by metabolic adaptations that suppress energy expenditure and facilitate weight regain. Skeletal muscle has a prominent role in energy homeostasis; therefore, we investigated the effect of WLM and weight regain on skeletal muscle in rodents. In skeletal muscle of obesity-prone rats, WLM reduced fat oxidative capacity and downregulated genes involved in fat metabolism. Interestingly, even after weight was regained, genes involved in fat metabolism were also reduced. We then subjected mice with skeletal muscle lipoprotein lipase overexpression (mCK-hLPL), which augments fat metabolism, to WLM and weight regain and found that mCK-hLPL attenuates weight regain by potentiating energy expenditure. Irrespective of genotype, weight regain suppressed dietary fat oxidation and downregulated genes involved in fat metabolism in skeletal muscle. However, mCK-hLPL mice oxidized more fat throughout weight regain and had greater expression of genes involved in fat metabolism and lower expression of genes involved in carbohydrate metabolism during WLM and regain. In summary, these results suggest that skeletal muscle fat oxidation is reduced during WLM and regain, and therapies that improve skeletal muscle fat metabolism may attenuate rapid weight regain.
Collapse
Affiliation(s)
- David M Presby
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Michael C Rudolph
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Vanessa D Sherk
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Matthew R Jackman
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Rebecca M Foright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS
| | - Kenneth L Jones
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Julie A Houck
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ginger C Johnson
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Janine A Higgins
- Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute and the Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Paul S MacLean
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
56
|
Shimizu Y, Tamura T, Kemmochi A, Owada Y, Ozawa Y, Hisakura K, Matsuzaka T, Shimano H, Nakano N, Sakashita S, Oda T, Ohkohchi N. Oxidative stress and Liver X Receptor agonist induce hepatocellular carcinoma in Non-alcoholic steatohepatitis model. J Gastroenterol Hepatol 2021; 36:800-810. [PMID: 32870526 PMCID: PMC7983938 DOI: 10.1111/jgh.15239] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM The incidence of non-alcoholic steatohepatitis (NASH)-related hepatocellular carcinoma (HCC) is progressively increasing. However, the pathophysiology and etiology of NASH progression to HCC are unknown. We hypothesized that steatosis was the key factor in NASH-related hepatocarcinogenesis and aimed to evaluate the effects of long-term liver X receptor (LXR) agonist stimulation on hepatic steatosis induced by a high-fat diet and oxidative stress. METHODS We used an LXR agonist (T0901317) and CCl4 to induce hepatic steatosis and oxidative stress, respectively. C57BL/6 mice fed with a high-fat diet were treated with either T0901317 + CCl4 (T09 + CCl4 group) or CCl4 alone (CCl4 group). T0901317 (2.5 mg/kg) and CCl4 (0.1 mL/kg) were intraperitoneally administered twice weekly for 24 weeks. RESULTS The liver-to-body weight ratio was significantly higher in the T09 + CCl4 group than in the CCl4 group. Mice in the T09 + CCl4 group exhibited abnormal lipid metabolism and NASH-like histopathological features. Additionally, all mice in the T09 + CCl4 group developed liver tumors diagnosed as well-differentiated HCC. The genes identified via microarray analysis were related to NASH and HCC development. CONCLUSIONS By combining long-term LXR agonist stimulation with oxidative stress and a high-fat diet, we successfully reproduced liver conditions in mice similar to those in humans with NASH and progression to HCC. Our results provide new insight into NASH-related HCC progression and therapy.
Collapse
Affiliation(s)
- Yoshio Shimizu
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Takafumi Tamura
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Akira Kemmochi
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yohei Owada
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Yusuke Ozawa
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Katsuji Hisakura
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Takashi Matsuzaka
- Department of Endocrinology and Metabolism, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Faculty of MedicineUniversity of TsukubaTsukubaJapan
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of TsukubaTsukubaJapan
- AMED‐CRESTJapan Agency for Medical Research and Development (AMED)TokyoJapan
| | - Noriyuki Nakano
- Department of Diagnostic Pathology, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Shingo Sakashita
- Department of Diagnostic Pathology, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Tatsuya Oda
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| | - Nobuhiro Ohkohchi
- Department of Gastrointestinal and Hepato‐Biliary‐Pancreatic Surgery, Faculty of MedicineUniversity of TsukubaTsukubaJapan
| |
Collapse
|
57
|
McKenna HT, O'Brien KA, Fernandez BO, Minnion M, Tod A, McNally BD, West JA, Griffin JL, Grocott MP, Mythen MG, Feelisch M, Murray AJ, Martin DS. Divergent trajectories of cellular bioenergetics, intermediary metabolism and systemic redox status in survivors and non-survivors of critical illness. Redox Biol 2021; 41:101907. [PMID: 33667994 PMCID: PMC7937570 DOI: 10.1016/j.redox.2021.101907] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/06/2021] [Accepted: 02/16/2021] [Indexed: 02/01/2023] Open
Abstract
Background Numerous pathologies result in multiple-organ failure, which is thought to be a direct consequence of compromised cellular bioenergetic status. Neither the nature of this phenotype nor its relevance to survival are well understood, limiting the efficacy of modern life-support. Methods To explore the hypothesis that survival from critical illness relates to changes in cellular bioenergetics, we combined assessment of mitochondrial respiration with metabolomic, lipidomic and redox profiling in skeletal muscle and blood, at multiple timepoints, in 21 critically ill patients and 12 reference patients. Results We demonstrate an end-organ cellular phenotype in critical illness, characterized by preserved total energetic capacity, greater coupling efficiency and selectively lower capacity for complex I and fatty acid oxidation (FAO)-supported respiration in skeletal muscle, compared to health. In survivors, complex I capacity at 48 h was 27% lower than in non-survivors (p = 0.01), but tended to increase by day 7, with no such recovery observed in non-survivors. By day 7, survivors’ FAO enzyme activity was double that of non-survivors (p = 0.048), in whom plasma triacylglycerol accumulated. Increases in both cellular oxidative stress and reductive drive were evident in early critical illness compared to health. Initially, non-survivors demonstrated greater plasma total antioxidant capacity but ultimately higher lipid peroxidation compared to survivors. These alterations were mirrored by greater levels of circulating total free thiol and nitrosated species, consistent with greater reductive stress and vascular inflammation, in non-survivors compared to survivors. In contrast, no clear differences in systemic inflammatory markers were observed between the two groups. Conclusion Critical illness is associated with rapid, specific and coordinated alterations in the cellular respiratory machinery, intermediary metabolism and redox response, with different trajectories in survivors and non-survivors. Unravelling the cellular and molecular foundation of human resilience may enable the development of more effective life-support strategies.
Collapse
Affiliation(s)
- Helen T McKenna
- Division of Surgery and Interventional Science, University College London, Royal Free Hospital, London, NW3 2QG, UK; Intensive Care Unit, Royal Free Hospital, London, NW3 2QG, UK; Peninsula Medical School, University of Plymouth, John Bull Building, Derriford, Plymouth, PL6 8BU, UK
| | - Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Bernadette O Fernandez
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Magdalena Minnion
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Adam Tod
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Ben D McNally
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, CB2 1GA, UK
| | - James A West
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, Jeffrey Cheah Biomedical Centre, University of Cambridge, CB2 0RE, UK
| | - Julian L Griffin
- Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, CB2 1GA, UK; Section of Biomolecular Medicine, Department of Digestion, Metabolism and Reproduction, Imperial College London, SW7 2AZ, UK
| | - Michael P Grocott
- Anaesthesia Perioperative and Critical Care Research Group, Southampton National Institute of Health Research Biomedical Research Centre, University Hospital Southampton, SO16 6YD, UK
| | - Michael G Mythen
- University College London Hospitals and Great Ormond Street, National Institute of Health Research Biomedical Research Centres, London, WC1N 1EH, UK
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK; Anaesthesia Perioperative and Critical Care Research Group, Southampton National Institute of Health Research Biomedical Research Centre, University Hospital Southampton, SO16 6YD, UK
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Daniel S Martin
- Division of Surgery and Interventional Science, University College London, Royal Free Hospital, London, NW3 2QG, UK; Intensive Care Unit, Royal Free Hospital, London, NW3 2QG, UK; Peninsula Medical School, University of Plymouth, John Bull Building, Derriford, Plymouth, PL6 8BU, UK
| |
Collapse
|
58
|
Virtual metabolic human dynamic model for pathological analysis and therapy design for diabetes. iScience 2021; 24:102101. [PMID: 33615200 PMCID: PMC7878987 DOI: 10.1016/j.isci.2021.102101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/21/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
A virtual metabolic human model is a valuable complement to experimental biology and clinical studies, because in vivo research involves serious ethical and technical problems. I have proposed a multi-organ and multi-scale kinetic model that formulates the reactions of enzymes and transporters with the regulation of hormonal actions at postprandial and postabsorptive states. The computational model consists of 202 ordinary differential equations for metabolites with 217 reaction rates and 1,140 kinetic parameter constants. It is the most comprehensive, largest, and highly predictive model of the whole-body metabolism. Use of the model revealed the mechanisms by which individual disorders, such as steatosis, β cell dysfunction, and insulin resistance, were combined to cause diabetes. The model predicted a glycerol kinase inhibitor to be an effective medicine for type 2 diabetes, which not only decreased hepatic triglyceride but also reduced plasma glucose. The model also enabled us to rationally design combination therapy. A standard of virtual metabolic human dynamic models is proposed It integrates the three scales of molecules, organs, and whole body It gets insight into pathological mechanisms of type 1 and type 2 diabetes It enables the computer-aided design of medication treatment for diabetes
Collapse
|
59
|
Pinto PR, Yoshinaga MY, Del Bianco V, Bochi AP, Ferreira GS, Pinto IFD, Rodrigues LG, Nakandakare ER, Okamoto MM, Machado UF, Miyamoto S, Catanozi S, Passarelli M. Dietary sodium restriction alters muscle lipidomics that relates to insulin resistance in mice. J Biol Chem 2021; 296:100344. [PMID: 33524391 PMCID: PMC7949138 DOI: 10.1016/j.jbc.2021.100344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
A low-sodium (LS) diet has been shown to reduce blood pressure (BP) and the incidence of cardiovascular diseases. However, severe dietary sodium restriction promotes insulin resistance (IR) and dyslipidemia in animal models and humans. Thus, further clarification of the long-term consequences of LS is needed. Here, we investigated the effects of chronic LS on gastrocnemius gene and protein expression and lipidomics and its association with IR and plasma lipids in LDL receptor knockout mice. Three-month-old male mice were fed a normal sodium diet (NS; 0.5% Na; n = 12-19) or LS (0.06% Na; n = 14-20) over 90 days. Body mass (BM), BP, plasma total cholesterol, triacylglycerol (TG), glucose, hematocrit, and IR were evaluated. LS increased BM (9%), plasma TG (51%), blood glucose (19%), and IR (46%) when compared with the NS. RT-qPCR analysis revealed that genes involved in lipid uptake and oxidation were increased by the LS: Fabp3 (106%), Prkaa1 (46%), and Cpt1 (74%). Genes and proteins (assessed by Western blotting) involved in insulin signaling were not changed by the LS. Similarly, lipid species classically involved in muscle IR, such as diacylglycerols and ceramides detected by ultra-high-performance liquid chromatography coupled to mass spectrometry, were also unchanged by LS. Species of phosphatidylcholines (68%), phosphatidylinositol (90%), and free fatty acids (59%) increased while cardiolipins (41%) and acylcarnitines (9%) decreased in gastrocnemius in response to LS and were associated with glucose disposal rate. Together these results suggest that chronic LS alters glycerophospholipid and fatty acids species in gastrocnemius that may contribute to glucose and lipid homeostasis derangements in mice.
Collapse
Affiliation(s)
- Paula Ramos Pinto
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marcos Y Yoshinaga
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Vanessa Del Bianco
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Paula Bochi
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Guilherme S Ferreira
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Isabella F D Pinto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Letícia G Rodrigues
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Edna R Nakandakare
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maristela M Okamoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ubiratan F Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Sergio Catanozi
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Marisa Passarelli
- Laboratório de Lípides (LIM-10), Hospital das Clínicas (HCFMUSP) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil; Programa de Pós-Graduação em Medicina, Universidade Nove de Julho, São Paulo, Brazil.
| |
Collapse
|
60
|
Sandoval C, Askelson K, Lambo CA, Dunlap KA, Satterfield MC. Effect of maternal nutrient restriction on expression of glucose transporters (SLC2A4 and SLC2A1) and insulin signaling in skeletal muscle of SGA and Non-SGA sheep fetuses. Domest Anim Endocrinol 2021; 74:106556. [PMID: 33120168 DOI: 10.1016/j.domaniend.2020.106556] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/08/2023]
Abstract
Maternal nutrient restriction (NR) causes small for gestational age (SGA) offspring, which are at higher risk for accelerated postnatal growth and developing insulin resistance in adulthood. Skeletal muscle is essential for whole-body glucose metabolism, as 80% of insulin-mediated glucose uptake occurs in this tissue. Maternal NR can alter fetal skeletal muscle mass, expression of glucose transporters, insulin signaling, and myofiber type composition. It also leads to accumulation of intramuscular triglycerides (IMTG), which correlates to insulin resistance. Using a 50% NR treatment from gestational day (GD) 35 to GD 135 in sheep, we routinely observe a spectral phenotype of fetal weights within the NR group. Thus, we classified those fetuses into NR(Non-SGA; n = 11) and NR(SGA; n = 11). The control group (n = 12) received 100% of nutrient requirements throughout pregnancy. At GD 135, fetal plasma and gastrocnemius and soleus muscles were collected. In fetal plasma, total insulin was lower in NR(SGA) fetuses compared NR(Non-SGA) and control fetuses (P < 0.01), whereas total IGF-1 was lower in NR(SGA) fetuses compared with control fetuses (P < 0.05). Within gastrocnemius, protein expression of insulin receptor (INSRB; P < 0.05) and the glucose transporters, solute carrier family 2 member 1 and solute carrier family 2 member 4, was higher (P < 0.05) in NR(SGA) fetuses compared with NR(Non-SGA) fetuses; IGF-1 receptor protein was increased (P < 0.01) in NR(SGA) fetuses compared with control fetuses, and a lower (P < 0.01) proportion of type I myofibers (insulin sensitive and oxidative) was observed in SGA fetuses. For gastrocnemius muscle, the expression of lipoprotein lipase (LPL) messenger RNA (mRNA) was upregulated (P < 0.05) in both NR(SGA) and NR(Non-SGA) fetuses compared with control fetuses, whereas carnitine palmitoyltransferase 1B (CPT1B) mRNA was higher (P < 0.05) in NR(Non-SGA) fetuses compared with control fetuses, but there were no differences (P > 0.05) for protein levels of LPL or CPT1B. Within soleus, there were no differences (P > 0.05) for any characteristic except for the proportion of type I myofibers, which was lower (P < 0.05) in NR(SGA) fetuses compared with control fetuses. Accumulation of IMTG did not differ (P > 0.05) in gastrocnemius or soleus muscles. Collectively, the results indicate molecular differences between SGA and Non-SGA fetuses for most characteristics, suggesting that maternal NR induces a spectral phenotype for the metabolic programming of those fetuses.
Collapse
Affiliation(s)
- C Sandoval
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA; Instituto de Investigaciones Agropecuarias, Región de Magallanes y la Antártica Chilena, Punta Arenas 6212707, Chile
| | - K Askelson
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - C A Lambo
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - K A Dunlap
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA
| | - M C Satterfield
- Department of Animal Science, Texas A&M University, College Station, TX 77845, USA.
| |
Collapse
|
61
|
Chi-Cervera LA, Montalvo GI, Icaza-Chávez ME, Torres-Romero J, Arana-Argáez V, Ramírez-Camacho M, Lara-Riegos J. Clinical relevance of lipid panel and aminotransferases in the context of hepatic steatosis and fibrosis as measured by transient elastography (FibroScan®). J Med Biochem 2021; 40:60-66. [PMID: 33584141 PMCID: PMC7857856 DOI: 10.5937/jomb0-24689] [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: 12/27/2019] [Accepted: 04/11/2020] [Indexed: 12/12/2022] Open
Abstract
Background Nonalcoholic fatty liver disease (NAFLD) is one of the most common causes of chronic liver disease and is associated with various co-morbidities. Transient elastography (FibroScan®) is a non-invasive method to detect NAFLD using the controlled attenuation parameter (CAP). We aimed to evaluate the association of the lipid panel and aminotransferases concentrations with the presence or absence of steatosis and fibrosis. Methods One hundred and five patients with NAFLD were included. Hepatic steatosis was quantified by CAP (dB/m) and liver stiffness by Kilopascals (kPa), these values were then analyzed against patient lipid panel and serum concentrations of the liver enzymes aspartate aminotransferase (AST) and alanine aminotransferase (ALT). A correlation and multiple regression were used. Mann-Whitney U test was used as non-parametric analysis. Results We observed an association between hepatic steatosis and total cholesterol (B = 0.021, p = 0.038, Exp (B) = 1.021, I.C = 1.001-1.041) as well as serum triglycerides (B = 0.017, p = 0.006, Exp (B) = 1.018 and I.C = 1.005-1.030). Similarly, we found an association between significant hepatic fibrosis and lower concentrations of total cholesterol (B = -0.019, p = 0.005, Exp (B) = 0.982 I.C = 0.969-0.995) and elevated AST (B = 0.042, p = 3.25 × 10-4, Exp (B) = 1.043 I.C = 1.019-1.068) independent of age, gender and BMI. Conclusions Our results suggest that, total cholesterol and triglyceride concentrations positively correlate with hepatic steatosis while significant hepatic fibrosis is associated with lower total cholesterol and higher AST concentrations.
Collapse
Affiliation(s)
| | - Gordon Iaarah Montalvo
- StarMédica Hospital, Gastrointestinal and Liver Speciality Clinic, Mérida, Yucatán, México
| | | | - Julio Torres-Romero
- Universidad Autónoma de Yucatán, Facultad de Química, Biochemistry and Molecular Genetics Laboratory, Mérida, Yucatán, México
| | - Víctor Arana-Argáez
- Universidad Autónoma de Yucatán, Facultad de Química, Pharmacology Laboratory, México
| | - Mario Ramírez-Camacho
- Universidad Autónoma de Yucatán, Facultad de Química, Drug Information Center, México
| | - Julio Lara-Riegos
- Universidad Autónoma de Yucatán, Facultad de Química, Biochemistry and Molecular Genetics Laboratory, Mérida, Yucatán, México
| |
Collapse
|
62
|
Kenny LC, Thomas G, Poston L, Myers JE, Simpson NAB, McCarthy FP, Brown LW, Bond AE, Tuytten R, Baker PN, on behalf of the Screening for Pregnancy Endpoints Consortium. Prediction of preeclampsia risk in first time pregnant women: Metabolite biomarkers for a clinical test. PLoS One 2020; 15:e0244369. [PMID: 33370367 PMCID: PMC7769282 DOI: 10.1371/journal.pone.0244369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/08/2020] [Indexed: 11/19/2022] Open
Abstract
Preeclampsia remains a leading cause of maternal and perinatal morbidity and mortality. Accurate prediction of preeclampsia risk would enable more effective, risk-based prenatal care pathways. Current risk assessment algorithms depend on clinical risk factors largely unavailable for first-time pregnant women. Delivering accurate preeclampsia risk assessment to this cohort of women, therefore requires for novel biomarkers. Here, we evaluated the relevance of metabolite biomarker candidates for their selection into a prototype rapid, quantitative Liquid Chromatography-tandem Mass Spectrometry (LC-MS/MS) based clinical screening assay. First, a library of targeted LC-MS/MS assays for metabolite biomarker candidates was developed, using a medium-throughput translational metabolomics workflow, to verify biomarker potential in the Screening-for-Pregnancy-Endpoints (SCOPE, European branch) study. A variable pre-selection step was followed by the development of multivariable prediction models for pre-defined clinical use cases, i.e., prediction of preterm preeclampsia risk and of any preeclampsia risk. Within a large set of metabolite biomarker candidates, we confirmed the potential of dilinoleoyl-glycerol and heptadecanoyl-2-hydroxy-sn-glycero-3-phosphocholine to effectively complement Placental Growth Factor, an established preeclampsia biomarker, for the prediction of preeclampsia risk in first-time pregnancies without overt risk factors. These metabolites will be considered for integration in a prototype rapid, quantitative LC-MS/MS assay, and subsequent validation in an independent cohort.
Collapse
Affiliation(s)
- Louise C. Kenny
- Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Grégoire Thomas
- SQU4RE, Lokeren, Belgium
- Metabolomic Diagnostics, Cork, Ireland
| | - Lucilla Poston
- Department of Women and Children’s Health, King’s College London, London, United Kingdom
| | - Jenny E. Myers
- Maternal & Fetal Health Research Centre, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Nigel A. B. Simpson
- Department of Women’s and Children’s Health, University of Leeds, Leeds, United Kingdom
| | - Fergus P. McCarthy
- Irish Centre for Fetal and Neonatal Translational Research, University College Cork, Cork, Ireland
| | | | | | | | - Philip N. Baker
- College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | | |
Collapse
|
63
|
|
64
|
The NLRP3 inflammasome regulates adipose tissue metabolism. Biochem J 2020; 477:1089-1107. [PMID: 32202638 DOI: 10.1042/bcj20190472] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/27/2022]
Abstract
Adipose tissue regulates metabolic homeostasis by participating in endocrine and immune responses in addition to storing and releasing lipids from adipocytes. Obesity skews adipose tissue adipokine responses and degrades the coordination of adipocyte lipogenesis and lipolysis. These defects in adipose tissue metabolism can promote ectopic lipid deposition and inflammation in insulin-sensitive tissues such as skeletal muscle and liver. Sustained caloric excess can expand white adipose tissue to a point of maladaptation exacerbating both local and systemic inflammation. Multiple sources, instigators and propagators of adipose tissue inflammation occur during obesity. Cross-talk between professional immune cells (i.e. macrophages) and metabolic cells (i.e. adipocytes) promote adipose tissue inflammation during metabolic stress (i.e. metaflammation). Metabolic stress and endogenous danger signals can engage pathogen recognition receptors (PRRs) of the innate immune system thereby activating pro-inflammatory and stress pathways in adipose tissue. The Nod-like receptor protein 3 (NLRP3) inflammasome can act as a metabolic danger sensor to a wide range of pathogen- and damage-associated molecular patterns (PAMPs and DAMPs). Activation of the NLRP3 inflammasome facilitates caspase-1 dependent production of the pro-inflammatory cytokines IL-1β and IL-18. Activation of the NLRP3 inflammasome can promote inflammation and pyroptotic cell death, but caspase-1 is also involved in adipogenesis. This review discusses the role of the NLRP3 inflammasome in adipose tissue immunometabolism responses relevant to metabolic disease. Understanding the potential sources of NLRP3 activation and consequences of NLRP3 effectors may reveal therapeutic opportunities to break or fine-tune the connection between metabolism and inflammation in adipose tissue during obesity.
Collapse
|
65
|
Chen S, Sbuh N, Veedu RN. Antisense Oligonucleotides as Potential Therapeutics for Type 2 Diabetes. Nucleic Acid Ther 2020; 31:39-57. [PMID: 33026966 DOI: 10.1089/nat.2020.0891] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder characterized by persistent hyperglycemia resulting from inefficient signaling and insufficient production of insulin. Conventional management of T2D has largely relied on small molecule-based oral hypoglycemic medicines, which do not halt the progression of the disease due to limited efficacy and induce adverse effects as well. To this end, antisense oligonucleotide has attracted immense attention in developing antidiabetic agents because of their ability to downregulate the expression of disease-causing genes at the RNA and protein level. To date, seven antisense agents have been approved by the United States Food and Drug Administration for therapies of a variety of human maladies, including genetic disorders. Herein, we provide a comprehensive review of antisense molecules developed for suppressing the causative genes believed to be responsible for insulin resistance and hyperglycemia toward preventing and treating T2D.
Collapse
Affiliation(s)
- Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Nabayet Sbuh
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| | - Rakesh N Veedu
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Australia.,Perron Institute for Neurological and Translational Science, Perth, Australia
| |
Collapse
|
66
|
Field BC, Gordillo R, Scherer PE. The Role of Ceramides in Diabetes and Cardiovascular Disease Regulation of Ceramides by Adipokines. Front Endocrinol (Lausanne) 2020; 11:569250. [PMID: 33133017 PMCID: PMC7564167 DOI: 10.3389/fendo.2020.569250] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic dysfunction is intertwined with the pathophysiology of both diabetes and cardiovascular disease. Recently, one particular lipid class has been shown to influence the development and sustainment of these diseases: ceramides. As a subtype of sphingolipids, these species are particularly central to many sphingolipid pathways. Increased levels of ceramides are known to correlate with impaired cardiovascular and metabolic health. Furthermore, the interaction between ceramides and adipokines, most notably adiponectin and leptin, appears to play a role in the pathophysiology of these conditions. Adiponectin appears to counteract the detrimental effects of elevated ceramides, largely through activation of the ceramidase activity of its receptors. Elevated ceramides appear to worsen leptin resistance, which is an important phenomenon in the pathophysiology of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Bianca C. Field
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
67
|
Maruta H, Yamashita H. Acetic acid stimulates G-protein-coupled receptor GPR43 and induces intracellular calcium influx in L6 myotube cells. PLoS One 2020; 15:e0239428. [PMID: 32997697 PMCID: PMC7526932 DOI: 10.1371/journal.pone.0239428] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022] Open
Abstract
Short chain fatty acids (SCFAs) produced endogenously in the gut by bacterial fermentation of dietary fiber have been studied as nutrients that act as signaling molecules to activate G-protein coupled receptors (GPCRs) such as GPR41 and GPR43. GPR43 functioning involves the suppression of lipid accumulation and maintaining body energy homeostasis, and is activated by acetic acid or propionic acid. Previously, we reported that the orally administered acetic acid improves lipid metabolism in liver and skeletal muscles and suppresses obesity, thus improving glucose tolerance. Acetic acid stimulates AMP-activated protein kinase (AMPK) through its metabolic pathway in skeletal muscle cells. We hypothesized that acetic acid would stimulate GPR43 in skeletal muscle cells and has function in modulating gene expression related to muscle characteristics through its signal pathway. The objective of the current study was to clarify this effect of acetic acid. The GPR43 expression, observed in the differentiated myotube cells, was increased upon acetic acid treatment. Acetic acid induced the intracellular calcium influx in the cells and this induction was significantly inhibited by the GPR43-specific siRNA treatment. The calcineurin molecule is activated by calcium/calmodulin and is associated with proliferation of slow-twitch fibers. Calcineurin was activated by acetic acid treatment and inhibited by the concomitant treatment with GPR43-siRNA. Acetic acid induced nuclear localization of myocyte enhancer factor 2A (MEF2A), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), and nuclear factor of activated t cells c1 (NFATc1). However, these localizations were abolished by the treatment with GPR43-siRNA. It was concluded that acetic acid plays a role in the activation of GPR43 and involves the proliferation of slow-twitch fibers in L6 skeletal muscles through the calcium-signaling pathway caused by induction of intracellular calcium influx.
Collapse
Affiliation(s)
- Hitomi Maruta
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
| | - Hiromi Yamashita
- Department of Nutritional Science, Faculty of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
- Graduate School of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama, Japan
- * E-mail:
| |
Collapse
|
68
|
Zheng X, Ren B, Li X, Yan H, Xie Q, Liu H, Zhou J, Tian J, Huang K. Selenoprotein F knockout leads to glucose and lipid metabolism disorders in mice. J Biol Inorg Chem 2020; 25:1009-1022. [DOI: 10.1007/s00775-020-01821-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
|
69
|
Barchetta I, Cimini FA, Chiappetta C, Bertoccini L, Ceccarelli V, Capoccia D, Gaggini M, Di Cristofano C, Della Rocca C, Silecchia G, Leonetti F, Lenzi A, Gastaldelli A, Cavallo MG. Relationship between hepatic and systemic angiopoietin-like 3, hepatic Vitamin D receptor expression and NAFLD in obesity. Liver Int 2020; 40:2139-2147. [PMID: 32510837 DOI: 10.1111/liv.14554] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide and an independent risk factor for cardiovascular mortality. Angiopoietin-like proteins (ANGPTLs) are targets for vitamin D receptor (VDR)-mediated gene transcription and this axis may promote NAFLD. ANGPTL3 is a hepatokine which inhibits lipoprotein lipase and its experimentally induced inactivation reduces hepatosteatosis. Little is known on ANGPTL3 in human NAFLD and no data exist on its relationship with hepatic VDR/VD-related genes. The aim of this research was to investigate hepatic ANGPTLs and VDR/VD-related gene expression in human obesity in relation to NAFLD. METHODS We conducted a cross-sectional investigation on forty obese subjects with/without NAFLD. We evaluated hepatic ANGPTL3, ANGPTL4, ANGPTL8, LPL, VDR, CYP27A1 and CYP2R1 mRNA expression in liver biopsies by RT-PCR; VDR expression was further investigated by immunohistochemistry; circulating ANGPTL3 was measured by Milliplex assay. RESULTS Compared to non-NAFLD, NAFLD individuals had significantly higher hepatic VDR, ANGPTL3 and LPL expression. ANGPTL3 correlated with steatosis grade, LPL, VDR, CYP27A1 and CYP2R1 expression. Plasma ANGPTL3 concentrations were positively associated with clinical/histological markers of NAFLD/NASH and with hepatic ANGPTL3 expression. Greater hepatic VDR expression was the main determinant of hepatic ANGPTL3 after adjusting for multiple confounders. CONCLUSIONS Hepatic ANGPTL3 expression correlates with greater VDR expression, presence and severity of NAFLD and translates in increased circulating ANGPTL3, likely as a result of its modulation by up-regulated hepatic VDR in NAFLD. This study provides novel insights to potential mechanisms underlying ANGPTLs-mediated ectopic fat accumulation and NAFLD development in obesity.
Collapse
Affiliation(s)
- Ilaria Barchetta
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Flavia A Cimini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Caterina Chiappetta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Laura Bertoccini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Danila Capoccia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Melania Gaggini
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Claudio Di Cristofano
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Carlo Della Rocca
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Gianfranco Silecchia
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Frida Leonetti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, CNR, Pisa, Italy
| | - Maria G Cavallo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
70
|
Spitler KM, Davies BSJ. Aging and plasma triglyceride metabolism. J Lipid Res 2020; 61:1161-1167. [PMID: 32586846 DOI: 10.1194/jlr.r120000922] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Indexed: 12/16/2022] Open
Abstract
The risk for metabolic disease, including metabolic syndrome, insulin resistance, and diabetes, increases with age. Altered plasma TG metabolism and changes in fatty acid partitioning are also major contributors to metabolic disease. Plasma TG metabolism itself is altered by age in humans and rodents. As discussed in this review, the age-induced changes in human TG metabolism include increased plasma TG levels, reduced postprandial plasma TG clearance rates, reduced postheparin LPL activity, decreased adipose tissue lipolysis, and elevated ectopic fat deposition, all of which could potentially contribute to age-associated metabolic diseases. Similar observations have been made in aged rats. We highlight the limitations of currently available data and propose that mechanistic studies are needed to understand the extent to which age-induced alterations in TG metabolism contribute to metabolic disease. Such mechanistic insights could aid in therapeutic strategies for preventing or managing metabolic disease in older individuals.
Collapse
Affiliation(s)
- Kathryn M Spitler
- Department of Biochemistry, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Brandon S J Davies
- Department of Biochemistry, Fraternal Order of Eagles Diabetes Research Center, and Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| |
Collapse
|
71
|
Xiao B, Mao J, Sun B, Zhang W, Wang Y, Wang P, Ruan Z, Xi W, Li H, Zhou J, Lu Y, Ding Q, Wang X, Liu J, Yan J, Luo C, Shi X, Yang R, Xi X. Integrin β3 Deficiency Results in Hypertriglyceridemia via Disrupting LPL (Lipoprotein Lipase) Secretion. Arterioscler Thromb Vasc Biol 2020; 40:1296-1310. [PMID: 32237906 DOI: 10.1161/atvbaha.119.313191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Integrin β3 is implicated in numerous biological processes such as its relevance to blood triglyceride, yet whether β3 deficiency affects this metabolic process remains unknown. Approach and Results: We showed that the Chinese patients with β3-deficient Glanzmann thrombasthenia had a 2-fold higher serum triglyceride level together with a lower serum LPL (lipoprotein lipase) level than those with an αIIb deficiency or healthy subjects. The β3 knockout mice recapitulated these phenotypic features. The elevated plasma triglyceride level was due to impaired LPL-mediated triglyceride clearance caused by a disrupted LPL secretion. Further analysis revealed that β3 directly bound LPL via a juxtamembrane TIH (threonine isoleucine histidine)720-722 motif in its cytoplasmic domain and functioned as an adaptor protein by interacting with LPL and PKD (protein kinase D) to form the PKD/β3/LPL complex that is required for β3-mediated LPL secretion. Furthermore, the impaired triglyceride clearance in β3 knockout mice could be corrected by adeno-associated virus serotype 9 (AAV9)-mediated delivery of wild-type but not TIH720-722-mutated β3 genes. CONCLUSIONS This study reveals a hypertriglyceridemia in both β3-deficient Chinese patients and mice and provides novel insights into the molecular mechanisms of the significant roles of β3 in LPL secretion and triglyceride metabolism, drawing attention to the metabolic consequences in patients with β3-deficient Glanzmann thrombasthenia.
Collapse
Affiliation(s)
- Bing Xiao
- From the State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, China (B.X., X.X.)
| | - Jianhua Mao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.M., W.Z., Y.W., P.W., Z.R., X.X.)
| | - Boyang Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (B.S., H.L., R.Y.)
| | - Wei Zhang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.M., W.Z., Y.W., P.W., Z.R., X.X.)
| | - Yun Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.M., W.Z., Y.W., P.W., Z.R., X.X.)
| | - Pengran Wang
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.M., W.Z., Y.W., P.W., Z.R., X.X.)
| | - Zheng Ruan
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.M., W.Z., Y.W., P.W., Z.R., X.X.)
| | - Wenda Xi
- Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China (W.X.)
| | - Huiyuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (B.S., H.L., R.Y.)
| | - Jingyi Zhou
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China (J.Z., Y.L., Q.D., X.W.)
| | - Yide Lu
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China (J.Z., Y.L., Q.D., X.W.)
| | - Qiulan Ding
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China (J.Z., Y.L., Q.D., X.W.)
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China (J.Z., Y.L., Q.D., X.W.)
| | - Jingqiu Liu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China (J.L., C.L.)
| | - Jinsong Yan
- Department of Hematology, Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Second Hospital of Dalian Medical University, China (J.Y.)
| | - Cheng Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China (J.L., C.L.)
| | - Xiaofeng Shi
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, China (X.S.)
| | - Renchi Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Hematological Disorders, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China (B.S., H.L., R.Y.)
| | - Xiaodong Xi
- From the State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, China (B.X., X.X.).,State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (J.M., W.Z., Y.W., P.W., Z.R., X.X.)
| |
Collapse
|
72
|
Yao C, Tian W, Song J, Wang J. Antihyperlipidaemic effect of microencapsulated Lactobacillus plantarum LIP-1 on hyperlipidaemic rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2007-2017. [PMID: 31849068 DOI: 10.1002/jsfa.10218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Previous studies have shown that Lactobacillus plantarum LIP-1 (hereafter LIP-1) has an obvious hypolipidemic effect, and microencapsulated probiotics can ensure the strains live through the gastrointestinal tract. Although there has been much research on both preparation and assessment methods for probiotics microcapsules, most assessments were made in vitro and few were validated in vivo. In this study, the protective effect of microencapsulation and the possible hypolipidemic mechanisms of probiotic LIP-1 were evaluated in rats. Treatments included rats fed on a normal diet, a high-fat diet, and a high-fat diet with an intragastric supplement of either non-microencapsulated LIP-1 cells (NME LIP-1) or microencapsulated LIP-1 (ME LIP-1). Lipid metabolism indicators were measured during the experiment and following euthanasia. RESULTS Microencapsulation increased survival and colonization of LIP-1 in the colon. ME LIP-1 was superior to NME LIP-1 in reducing cholesterol. The mechanisms behind the hypolipidemic effect exerted by LIP-1 are possibly due to promoting the excretion of cholesterol, improving antioxygenic potentials, enhancing recovery from the injury in the liver, cardiovascular intima and intestinal mucosa, promoting the generation of short-chain fatty acids, and improving lipid metabolism. CONCLUSIONS This study confirms that microencapsulation provides effective protection of LIP-1 in the digestive system and the role of LIP-1 in the prevention and cure of hyperlipidaemia, providing theoretical support for probiotics to enter clinical applications. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Caiqing Yao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Wenjing Tian
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
- Department of Food and Biological Engineering, Beijing Vocational College of Agriculture, Beijing, China
| | - Jiaojiao Song
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| | - Junguo Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
73
|
Cirera S, Taşöz E, Juul Jacobsen M, Schumacher-Petersen C, Østergaard Christoffersen B, Kaae Kirk R, Pagh Ludvigsen T, Hvid H, Duelund Pedersen H, Høier Olsen L, Fredholm M. The expression signatures in liver and adipose tissue from obese Göttingen Minipigs reveal a predisposition for healthy fat accumulation. Nutr Diabetes 2020; 10:9. [PMID: 32205840 PMCID: PMC7090036 DOI: 10.1038/s41387-020-0112-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Model animals are valuable resources for dissecting basic aspects of the regulation of obesity and metabolism. The translatability of results relies on understanding comparative aspects of molecular pathophysiology. Several studies have shown that despite the presence of overt obesity and dyslipidemia in the pig key human pathological hepatic findings such as hepatocellular ballooning and abundant steatosis are lacking in the model. OBJECTIVES The aim of this study was to elucidate why these histopathological characteristics did not occur in a high fat, fructose and cholesterol (FFC) diet-induced obese Göttingen Minipig model. METHODS High-throughput expression profiling of more than 90 metabolically relevant genes was performed in liver, subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) of male minipigs diet fed: standard chow (SD, n = 7); FFC diet (n = 14); FFC diet in streptozotocin-induced diabetic pigs (FFCDIA, n = 8). Moreover, histopathological assessment of SAT and VAT was performed. RESULTS 12, 4 and 1 genes were highly significantly differentially expressed in liver, SAT and VAT when comparing the FFC and SD groups whereas the corresponding numbers were 15, 2, and 1 when comparing the FFCDIA and SD groups. Although the minipigs in both FFC groups developed sever obesity and dyslipidemia, the insulin-signaling pathways were not affected. Notably, four genes involved in lipid acquisition and removal, were highly deregulated in the liver: PPARG, LPL, CD36 and FABP4. These genes have been reported to play a major role in promoting hepatic steatosis in rodents and humans. Since very little macrophage-associated pro-inflammatory response was detected in the adipose tissues the expansion appears to have no adverse impact on adipose tissue metabolism. CONCLUSION The study shows that morbidly obese Göttingen Minipigs are protected against many of the metabolic and hepatic abnormalities associated with obesity due to a remarkable ability to expand the adipose compartments to accommodate excess calories.
Collapse
Affiliation(s)
- Susanna Cirera
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Emirhan Taşöz
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Mette Juul Jacobsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Camilla Schumacher-Petersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | | | - Rikke Kaae Kirk
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | | | - Henning Hvid
- Global Drug Discovery, Novo Nordisk A/S, Novo Nordisk Park, Måløv, Denmark
| | - Henrik Duelund Pedersen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
- Ellegaard Gottingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Lisbeth Høier Olsen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark
| | - Merete Fredholm
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 1870, Frederiksberg, Denmark.
| |
Collapse
|
74
|
Lake JA, Abasht B. Glucolipotoxicity: A Proposed Etiology for Wooden Breast and Related Myopathies in Commercial Broiler Chickens. Front Physiol 2020; 11:169. [PMID: 32231585 PMCID: PMC7083144 DOI: 10.3389/fphys.2020.00169] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/13/2020] [Indexed: 01/10/2023] Open
Abstract
Wooden breast is one of several myopathies of fast-growing commercial broilers that has emerged as a consequence of intensive selection practices in the poultry breeding industry. Despite the substantial economic burden presented to broiler producers worldwide by wooden breast and related muscle disorders such as white striping, the genetic and etiological underpinnings of these diseases are still poorly understood. Here we propose a new hypothesis on the primary causes of wooden breast that implicates dysregulation of lipid and glucose metabolism. Our hypothesis addresses recent findings that have suggested etiologic similarities between wooden breast and type 2 diabetes despite their phenotypic disparities. Unlike in mammals, dysregulation of lipid and glucose metabolism is not accompanied by an increase in plasma glucose levels but generates a unique skeletal muscle phenotype, i.e., wooden breast, in chickens. We hypothesize that these phenotypic disparities result from a major difference in skeletal muscle glucose transport between birds and mammals, and that the wooden breast phenotype most closely resembles complications of diabetes in smooth and cardiac muscle of mammals. Additional basic research on wooden breast and related muscle disorders in commercial broiler chickens is necessary and can be informative for poultry breeding and production as well as for human health and disease. To inform future studies, this paper reviews the current biological knowledge of wooden breast, outlines the major steps in its proposed pathogenesis, and examines how selection for production traits may have contributed to its prevalence.
Collapse
Affiliation(s)
- Juniper A. Lake
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, United States
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, United States
| | - Behnam Abasht
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, United States
- Department of Animal and Food Sciences, University of Delaware, Newark, DE, United States
| |
Collapse
|
75
|
Zhang B, Zhang C, Zhang X, Li N, Dong Z, Sun G, Sun X. Atorvastatin promotes AMPK signaling to protect against high fat diet-induced non-alcoholic fatty liver in golden hamsters. Exp Ther Med 2020; 19:2133-2142. [PMID: 32104276 PMCID: PMC7027324 DOI: 10.3892/etm.2020.8465] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 12/11/2019] [Indexed: 12/19/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by diffuse fatty acid degeneration and excess fat accumulation in the liver. Notably, the currently available medications used to treat NAFLD remain limited. The aim of the present study was to investigate the protective role of atorvastatin (Ato) against NAFLD in golden hamsters fed a high fat diet (HFD) and in HepG2 cells treated with palmitate, and identify the underlying molecular mechanism. Ato (3 mg/kg) was administered orally every day for 8 weeks to the hamsters during HFD administration. Hamsters in the model group developed hepatic steatosis with high serum levels of triglyceride, cholesterol, insulin and C-reactive protein, which were effectively reduced by treatment with Ato. Additionally, the relative liver weight of hamsters treated with Ato was markedly lower compared with that of the model group. Hematoxylin and eosin, and oil red O staining indicated that the livers of the animals in the model group exhibited large and numerous lipid droplets, which were markedly decreased after Ato treatment. Western blot analysis indicated that Ato inhibited fat accumulation in the liver through the AMP-activated protein kinase (AMPK)-dependent activation of peroxisome proliferator activated receptor α (PPARα), peroxisome proliferator-activated receptor-γ coactivator 1 α and their target genes. Furthermore, in vitro, Ato inhibited PA-induced lipid accumulation in HepG2 cells. This inhibitory effect was attenuated following Compound C treatment, indicating that AMPK may be a potential target of Ato. In conclusion, the increase in AMPK-mediated PPARα and its target genes may represent a novel molecular mechanism by which Ato prevents NAFLD.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, P.R. China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, P.R. China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, P.R. China
| | - Chenyang Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, P.R. China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, P.R. China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, P.R. China
| | - Xuelian Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, P.R. China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, P.R. China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, P.R. China
| | - Nannan Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| | - Zhengqi Dong
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, P.R. China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, P.R. China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, P.R. China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, P.R. China.,Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, P.R. China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, P.R. China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, P.R. China
| |
Collapse
|
76
|
Fan W, Liu W, Liu H, Meng Q, Xu Y, Guo Y, Wang B, Zhou Z, Hou S. Dynamic accumulation of fatty acids in duck (Anas platyrhynchos) breast muscle and its correlations with gene expression. BMC Genomics 2020; 21:58. [PMID: 31952469 PMCID: PMC6969424 DOI: 10.1186/s12864-020-6482-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Background Fatty acid composition contributes greatly to the quality and nutritional value of meat. However, the molecular regulatory mechanisms underlying fatty acid accumulation in poultry have not yet been cleared. The aims of this study were to characterize the dynamics of fatty acid accumulation in duck breast muscle and investigate its correlations with gene expression. Results Here, we analyzed the fatty acid profile and transcriptome of breast muscle derived from Pekin ducks and mallards at the ages of 2 weeks, 4 weeks, 6 weeks and 8 weeks. Twenty fatty acids were detected in duck breast muscle, with palmitic acid (C16:0, 16.6%~ 21.1%), stearic acid (C18:0, 9.8%~ 17.7%), oleic acid (C18:1n-9, 15.7%~ 33.8%), linoleic acid (C18:2n-6, 10.8%~ 18.9%) and arachidonic acid (C20:4n-6, 11.7%~ 28.9%) as the major fatty acids. Our results showed that fatty acid composition was similar between the two breeds before 6 weeks, but the compositions diverged greatly after this point, mainly due to the stronger capacity for C16:0 and C18:1n-9 deposition in Pekin ducks. By comparing the multistage transcriptomes of Pekin ducks and mallards, we identified 2025 differentially expressed genes (DEGs). Cluster analysis of these DEGs revealed that the genes involved in oxidative phosphorylation, fatty acid degradation and the PPAR signaling pathway were upregulated in mallard at 8 weeks. Moreover, correlation analysis of the DEGs and fatty acid composition traits suggested that the DEGs involved in lipogenesis, lipolysis and fatty acid β-oxidation may interact to influence the deposition of fatty acids in duck breast muscle. Conclusions We reported the temporal progression of fatty acid accumulation and the dynamics of the transcriptome in breast muscle of Pekin ducks and mallards. Our results provide insights into the transcriptome regulation of fatty acid accumulation in duck breast muscle, and will facilitate improvements of fatty acid composition in duck breeding.
Collapse
Affiliation(s)
- Wenlei Fan
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan W Rd, Beijing, 100193, China.,College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China.,College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Wenjing Liu
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Hehe Liu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan W Rd, Beijing, 100193, China
| | - Qingshi Meng
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan W Rd, Beijing, 100193, China
| | - Yaxi Xu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan W Rd, Beijing, 100193, China
| | - Yuming Guo
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People's Republic of China
| | - Baowei Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, People's Republic of China
| | - Zhengkui Zhou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan W Rd, Beijing, 100193, China.
| | - Shuisheng Hou
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs; State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, No. 2 Yuanmingyuan W Rd, Beijing, 100193, China.
| |
Collapse
|
77
|
Jang K, Tong T, Lee J, Park T, Lee H. Altered Gene Expression Profiles in Peripheral Blood Mononuclear Cells in Obese Subjects. Obes Facts 2020; 13:375-385. [PMID: 32544907 PMCID: PMC7445570 DOI: 10.1159/000507817] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Gene expression profiles in human peripheral blood mononuclear cells (PBMCs) may act as a useful tool to better understand obesity. We investigated gene expression levels in PMBCs for possible differences between obese and non-obese subjects (19-55 years) and evaluated correlations between gene expression in PBMCs and clinical obesity indices. METHODS Body weight, BMI, fat amount, fat percentage, waist/hip ratio, leptin, and adiponectin levels were determined in 30 obese and 20 non-obese subjects. Expression levels of 19 genes, which were differentially expressed by clinical obesity indices in the PBMCs of high fat-fed rats, were determined in their PBMCs using real-time PCR. RESULTS The expression of 9 of 19 previously selected genes was significantly correlated with one or more clinical obesity indices. Both TFEC and CCL2 expression were negatively correlated with BMI, fat amount, fat percentage, waist/hip ratio, and leptin concentration. Similarly, TNFAIP2, VCAN, ASSI, IRF1, and HK3 expression negatively correlated with some clinical obesity indices, such as TNFAIP2 for BMI, fat amount, fat percentage, and waist/hip ratio, VCAN for fat amount, fat percentage, and waist/hip ratio, ASS1 for BMI and fat amount, IRF1 for BMI, fat amount, and fat percentage, and HK3 for fat amount. In contrast, both TNF-α and LPL expression were positively correlated with waist/hip ratio. CONCLUSION We identified 9 of 19 genes in human PBMCs that significantly correlated with one or more clinical obesity indices. Because these genes have a mechanistic basis for the development or progression of obesity and its metabolic derangements, they may help to determine possible underlying mechanisms for obesity.
Collapse
Affiliation(s)
- Kyungho Jang
- Center for Clinical Pharmacology, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Republic of Korea
| | - Tao Tong
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Jinhui Lee
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, Yonsei University, Seoul, Republic of Korea
| | - Taesun Park
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, Yonsei University, Seoul, Republic of Korea
- **Taesun Park, Department of Food and Nutrition, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120749 (South Korea),
| | - Howard Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
- *Howard Lee, Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, 103 Daehak-ro, Jongno-gu, Seoul 110799 (South Korea),
| |
Collapse
|
78
|
Akhtar DH, Iqbal U, Vazquez-Montesino LM, Dennis BB, Ahmed A. Pathogenesis of Insulin Resistance and Atherogenic Dyslipidemia in Nonalcoholic Fatty Liver Disease. J Clin Transl Hepatol 2019; 7:362-370. [PMID: 31915606 PMCID: PMC6943204 DOI: 10.14218/jcth.2019.00028] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/08/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the developed world, with a global prevalence of around 25%. NAFLD is considered to be the hepatic manifestation of metabolic syndrome and is strongly associated with obesity, insulin resistance and dyslipidemia. Insulin resistance plays a pivotal role in the development of NAFLD-related dyslipidemia, which ultimately increases the risk of premature cardiovascular diseases, a leading cause of morbidity and mortality in patients with NAFLD. Insulin affects hepatic glucose and lipid metabolism by hepatic or extrahepatic pathways. Aside from insulin resistance, several other factors also contribute to the pathogenesis of atherogenic dyslipidemia in patients with NAFLD. These include diet composition, gut microbiota and genetic factors, to name a few. The identification of potentially modifiable risk factors of NAFLD is of importance, so as to target those who may benefit from lifestyle changes and to help develop targeted therapies that decrease the risk of cardiovascular diseases in patients with NAFLD.
Collapse
Affiliation(s)
- Daud H. Akhtar
- Department of Medicine, University of British Columbia Faculty of Medicine, Vancouver BC, Canada
| | - Umair Iqbal
- Department of Medicine, Geisinger Commonwealth School of Medicine, Danville, PA, USA
- *Correspondence to: Umair Iqbal, Department of Medicine, Geisinger Commonwealth School of Medicine, Danville, PA 17821, USA. Tel: +1-570-271-6211, E-mail:
| | | | - Brittany B. Dennis
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton ON, Canada
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aijaz Ahmed
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
79
|
Christodoulou MI, Tchoumtchoua J, Skaltsounis AL, Scorilas A, Halabalaki M. Natural Alkaloids Intervening the Insulin Pathway: New Hopes for Anti-Diabetic Agents? Curr Med Chem 2019; 26:5982-6015. [PMID: 29714135 DOI: 10.2174/0929867325666180430152618] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/16/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Accumulating experimental data supports the capacity of natural compounds to intervene in complicated molecular pathways underlying the pathogenesis of certain human morbidities. Among them, diabetes is now a world's epidemic associated with increased risk of death; thus, the detection of novel anti-diabetic agents and/or adjuvants is of vital importance. Alkaloids represent a diverse group of natural products with a range of therapeutic properties; during the last 20 years, published research on their anti-diabetic capacity has been tremendously increased. PURPOSE To discuss current concepts on the anti-diabetic impact of certain alkaloids, with special reference to their molecular targets throughout the insulin-signaling pathway. METHODOLOGY Upon in-depth search in the SCOPUS and PUBMED databases, the literature on alkaloids with insulin secretion/sensitization properties was critically reviewed. RESULTS In-vitro and in-vivo evidence supports the effect of berberine, trigonelline, piperine, oxymatrine, vindoneline, evodiamine and neferine on insulin-signaling and related cascades in beta-cells, myocytes, adipocytes, hepatocytes and other cells. Associated receptors, kinases, hormones and cytokines, are affected in terms of gene transcription, protein expression, activity and/or phosphorylation. Pathophysiological processes associated with insulin resistance, beta-cell failure, oxidative stress and inflammation, as well as clinical phenotype are also influenced. DISCUSSION Growing evidence suggests the ability of specific alkaloids to intervene in the insulin-signal transduction pathway, reverse molecular defects resulting in insulin resistance and glucose intolerance and improve disease complications, in-vitro and in-vivo. Future indepth molecular studies are expected to elucidate their exact mechanism of action, while large clinical trials are urgently needed to assess their potential as anti-diabetic agents.
Collapse
Affiliation(s)
- Maria-Ioanna Christodoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimioupoli Zografou 15771, Athens, Greece
| | - Job Tchoumtchoua
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupoli Zografou 15771, Athens, Greece
| | - Alexios-Leandros Skaltsounis
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupoli Zografou 15771, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimioupoli Zografou 15771, Athens, Greece
| | - Maria Halabalaki
- Department of Pharmacognosy and Natural Product Chemistry, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupoli Zografou 15771, Athens, Greece
| |
Collapse
|
80
|
Lecoutre S, Montel V, Vallez E, Pourpe C, Delmont A, Eury E, Verbanck M, Dickes-Coopman A, Daubersies P, Lesage J, Laborie C, Tailleux A, Staels B, Froguel P, Breton C, Vieau D. Transcription profiling in the liver of undernourished male rat offspring reveals altered lipid metabolism pathways and predisposition to hepatic steatosis. Am J Physiol Endocrinol Metab 2019; 317:E1094-E1107. [PMID: 31638854 DOI: 10.1152/ajpendo.00291.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Clinical and animal studies have reported an association between low birth weight and the development of nonalcoholic fatty liver disease (NAFLD) in offspring. Using a model of prenatal maternal 70% food restriction diet (FR30) in the rat, we previously showed that maternal undernutrition predisposes offspring to altered lipid metabolism in adipose tissue, especially on a high-fat (HF) diet. Here, using microarray-based expression profiling combined with metabolic, endocrine, biochemical, histological, and lipidomic approaches, we assessed whether FR30 procedure sensitizes adult male offspring to impaired lipid metabolism in the liver. No obvious differences were noted in the concentrations of triglycerides, cholesterol, and bile acids in the liver of 4-mo-old FR30 rats whichever postweaning diet was used. However, several clues suggest that offspring's lipid metabolism and steatosis are modified by maternal undernutrition. First, lipid composition was changed (i.e., higher total saturated fatty acids and lower elaidic acid) in the liver, whereas larger triglyceride droplets were observed in hepatocytes of undernourished rats. Second, FR30 offspring exhibited long-term impact on hepatic gene expression and lipid metabolism pathways on a chow diet. Although the transcriptome profile was globally modified by maternal undernutrition, cholesterol and bile acid biosynthesis pathways appear to be key targets, indicating that FR30 animals were predisposed to impaired hepatic cholesterol metabolism. Third, the FR30 protocol markedly modifies hepatic gene transcription profiles in undernourished offspring in response to postweaning HF. Overall, FR30 offspring may exhibit impaired metabolic flexibility, which does not enable them to properly cope with postweaning nutritional challenges influencing the development of nonalcoholic fatty liver.
Collapse
Affiliation(s)
- Simon Lecoutre
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Valérie Montel
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Emmanuelle Vallez
- Université Lille, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, INSERM U1011-European Genomic Institute for Diabetes, Lille, France
| | - Charlène Pourpe
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | | | - Elodie Eury
- Université Lille, UMR 8199, European Genomic Institute for Diabetes, Lille, France
| | - Marie Verbanck
- Université Lille, UMR 8199, European Genomic Institute for Diabetes, Lille, France
| | - Anne Dickes-Coopman
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | | | - Jean Lesage
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Christine Laborie
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Anne Tailleux
- Université Lille, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, INSERM U1011-European Genomic Institute for Diabetes, Lille, France
| | - Bart Staels
- Université Lille, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, INSERM U1011-European Genomic Institute for Diabetes, Lille, France
| | - Philippe Froguel
- Université Lille, UMR 8199, European Genomic Institute for Diabetes, Lille, France
| | - Christophe Breton
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| | - Didier Vieau
- Université Lille, EA4489, Equipe Malnutrition Maternelle et Programmation des Maladies Métaboliques, Lille, France
| |
Collapse
|
81
|
Di Pino A, DeFronzo RA. Insulin Resistance and Atherosclerosis: Implications for Insulin-Sensitizing Agents. Endocr Rev 2019; 40:1447-1467. [PMID: 31050706 PMCID: PMC7445419 DOI: 10.1210/er.2018-00141] [Citation(s) in RCA: 276] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022]
Abstract
Patients with type 2 diabetes mellitus (T2DM) are at high risk for macrovascular complications, which represent the major cause of mortality. Despite effective treatment of established cardiovascular (CV) risk factors (dyslipidemia, hypertension, procoagulant state), there remains a significant amount of unexplained CV risk. Insulin resistance is associated with a cluster of cardiometabolic risk factors known collectively as the insulin resistance (metabolic) syndrome (IRS). Considerable evidence, reviewed herein, suggests that insulin resistance and the IRS contribute to this unexplained CV risk in patients with T2DM. Accordingly, CV outcome trials with pioglitazone have demonstrated that this insulin-sensitizing thiazolidinedione reduces CV events in high-risk patients with T2DM. In this review the roles of insulin resistance and the IRS in the development of atherosclerotic CV disease and the impact of the insulin-sensitizing agents and of other antihyperglycemic medications on CV outcomes are discussed.
Collapse
Affiliation(s)
- Antonino Di Pino
- Diabetes Division, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas
| | - Ralph A DeFronzo
- Diabetes Division, University of Texas Health Science Center and Texas Diabetes Institute, San Antonio, Texas
| |
Collapse
|
82
|
Vorotnikov AV, Stafeev IS, Menshikov MY, Shestakova MV, Parfyonova YV. Latent Inflammation and Defect in Adipocyte Renewal as a Mechanism of Obesity-Associated Insulin Resistance. BIOCHEMISTRY (MOSCOW) 2019; 84:1329-1345. [DOI: 10.1134/s0006297919110099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
83
|
Vazquez-Gomez M, Heras-Molina A, Garcia-Contreras C, Pesantez-Pacheco JL, Torres-Rovira L, Martinez-Fernandez B, Gonzalez J, Encinas T, Astiz S, Ovilo C, Isabel B, Gonzalez-Bulnes A. Polyphenols and IUGR Pregnancies: Effects of Maternal Hydroxytyrosol Supplementation on Postnatal Growth, Metabolism and Body Composition of the Offspring. Antioxidants (Basel) 2019; 8:E535. [PMID: 31717349 PMCID: PMC6912388 DOI: 10.3390/antiox8110535] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/28/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
Maternal supplementation with the polyphenol hydroxytyrosol in a swine model of intrauterine growth restriction (IUGR) improves the fetal oxidative status, decreases the appearance of low birth-weight neonates and favors growth during early postnatal stages (lactation). The current study aimed to determine whether hydroxytyrosol supplementation can also improve developmental patterns, metabolic traits, and body composition of the offspring during later postnatal stages (from weaning to adulthood). A total of 21 piglets born from control untreated sows and 20 piglets born from sows treated with hydroxytyrosol during the last two-thirds of pregnancy were selected on the basis of similar body weights at weaning, for avoiding any interfering effects occurred during lactation. The pigs in the treated group had higher average daily weight gain (ADWG) and, therefore, reached higher body weight and corpulence, greater muscle development and higher adiposity than their control counterparts. The following were not found: significant effects on metabolism and body composition except changes in the muscular fatty acid composition of the treated pigs coming from the largest litters; those more affected by IUGR processes. These findings suggest that maternal supplementation with hydroxytyrosol may improve juvenile development of offspring in at-risk pregnancies and pave the way for more specific studies aiming to elucidate effects on adiposity, metabolism, and meat organoleptic characteristics.
Collapse
Affiliation(s)
- Marta Vazquez-Gomez
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n. 28040 Madrid, Spain; (M.V.-G.); (T.E.); (B.I.)
| | - Ana Heras-Molina
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
| | - Consolacion Garcia-Contreras
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
| | - Jose Luis Pesantez-Pacheco
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
- School of Veterinary Medicine and Zootechnics, Faculty of Agricultural Sciences, University of Cuenca, Avda. Doce de Octubre, 010220 Cuenca, Ecuador
| | - Laura Torres-Rovira
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
| | | | - Jorge Gonzalez
- Micros Veterinaria, Campus de Vegazana, 24007 Leon, Spain; (B.M.-F.); (J.G.)
| | - Teresa Encinas
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n. 28040 Madrid, Spain; (M.V.-G.); (T.E.); (B.I.)
| | - Susana Astiz
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
| | - Cristina Ovilo
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
| | - Beatriz Isabel
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n. 28040 Madrid, Spain; (M.V.-G.); (T.E.); (B.I.)
| | - Antonio Gonzalez-Bulnes
- Faculty of Veterinary Medicine, UCM, Ciudad Universitaria s/n. 28040 Madrid, Spain; (M.V.-G.); (T.E.); (B.I.)
- SGIT-INIA, Ctra. de La Coruña Km. 7,5. 29040 Madrid, Spain; (A.H.-M.); (C.G.-C.); (J.L.P.-P.); (L.T.-R.); (S.A.); (C.O.)
| |
Collapse
|
84
|
Singh RG, Nguyen NN, Cervantes A, Alarcon Ramos GC, Cho J, Petrov MS. Associations between intra-pancreatic fat deposition and circulating levels of cytokines. Cytokine 2019; 120:107-114. [DOI: 10.1016/j.cyto.2019.04.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/29/2019] [Accepted: 04/18/2019] [Indexed: 12/12/2022]
|
85
|
Wang T, Xiao X, Regenstein JM, Wu W, Zhou Y, Wang S, Cheng Y, Wu X, Bao B. Effect on lipid metabolism of mice continuously fed a crab-containing diet. FOOD BIOSCI 2019. [DOI: 10.1016/j.fbio.2019.100422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
86
|
Insulin Sensitivity Is Associated with Lipoprotein Lipase ( LPL) and Catenin Delta 2 ( CTNND2) DNA Methylation in Peripheral White Blood Cells in Non-Diabetic Young Women. Int J Mol Sci 2019; 20:ijms20122928. [PMID: 31208038 PMCID: PMC6627674 DOI: 10.3390/ijms20122928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/03/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Hyperglycaemia and type 2 diabetes (T2D) are associated with impaired insulin secretion and/or insulin action. Since few studies have addressed the relation between DNA methylation patterns with elaborated surrogates of insulin secretion/sensitivity based on the intravenous glucose tolerance test (IVGTT), the aim of this study was to evaluate the association between DNA methylation and an insulin sensitivity index based on IVGTT (calculated insulin sensitivity index (CSi)) in peripheral white blood cells from 57 non-diabetic female volunteers. The CSi and acute insulin response (AIR) indexes, as well as the disposition index (DI = CSi × AIR), were estimated from abbreviated IVGTT in 49 apparently healthy Chilean women. Methylation levels were assessed using the Illumina Infinium Human Methylation 450k BeadChip. After a statistical probe filtering, the two top CpGs whose methylation was associated with CSi were cg04615668 and cg07263235, located in the catenin delta 2 (CTNND2) and lipoprotein lipase (LPL) genes, respectively. Both CpGs conjointly predicted insulin sensitivity status with an area under the curve of 0.90. Additionally, cg04615668 correlated with homeostasis model assessment insulin-sensitivity (HOMA-S) and AIR, whereas cg07263235 was associated with plasma creatinine and DI. These results add further insights into the epigenetic regulation of insulin sensitivity and associated complications, pointing the CTNND2 and LPL genes as potential underlying epigenetic biomarkers for future risk of insulin-related diseases.
Collapse
|
87
|
Teratani T, Tomita K, Furuhashi H, Sugihara N, Higashiyama M, Nishikawa M, Irie R, Takajo T, Wada A, Horiuchi K, Inaba K, Hanawa Y, Shibuya N, Okada Y, Kurihara C, Nishii S, Mizoguchi A, Hozumi H, Watanabe C, Komoto S, Nagao S, Yamamoto J, Miura S, Hokari R, Kanai T. Lipoprotein Lipase Up-regulation in Hepatic Stellate Cells Exacerbates Liver Fibrosis in Nonalcoholic Steatohepatitis in Mice. Hepatol Commun 2019; 3:1098-1112. [PMID: 31388630 PMCID: PMC6671781 DOI: 10.1002/hep4.1383] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022] Open
Abstract
Lipoprotein lipase (LPL) plays a central role in incorporating plasma lipids into tissues and regulates lipid metabolism and energy balance in the human body. Conversely, LPL expression is almost absent in normal adult livers. Therefore, its physiological role in the liver remains unknown. We aimed to elucidate the role of LPL in the pathophysiology of nonalcoholic steatohepatitis (NASH), a hepatic manifestation of obesity. Hepatic stellate cell (HSC)–specific LPL‐knockout (LplHSC‐KO) mice, LPL‐floxed (Lplfl/fl) mice, or double‐mutant toll‐like receptor 4–deficient (Tlr4−/−) LplHSC‐KO mice were fed a high‐fat/high‐cholesterol diet for 4 weeks to establish the nonalcoholic fatty liver model or an high‐fat/high‐cholesterol diet for 24 weeks to establish the NASH model. Human samples, derived from patients with nonalcoholic fatty liver disease, were also examined. In human and mouse NASH livers, serum obesity‐related factors, such as free fatty acid, leptin, and interleukin‐6, dramatically increased the expression of LPL, specifically in HSCs through signal transducer and activator of transcription 3 signaling, as opposed to that in hepatocytes or hepatic macrophages. In the NASH mouse model, liver fibrosis was significantly reduced in LplHSC‐KO mice compared with that in Lplfl/fl mice. Nonenzymatic LPL‐mediated cholesterol uptake from serum lipoproteins enhanced the accumulation of free cholesterol in HSCs, which amplified TLR4 signaling, resulting in the activation of HSCs and progression of hepatic fibrosis in NASH. Conclusion: The present study reveals the pathophysiological role of LPL in the liver, and furthermore, clarifies the pathophysiology in which obesity, as a background factor, exacerbates NASH. The LPL‐mediated HSC activation pathway could be a promising therapeutic target for treating liver fibrosis in NASH.
Collapse
Affiliation(s)
- Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine Keio University School of Medicine Shinjuku-ku Tokyo Japan
| | - Kengo Tomita
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Hirotaka Furuhashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Nao Sugihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Masaaki Higashiyama
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Makoto Nishikawa
- Department of Surgery National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Rie Irie
- Department of Pathology National Center for Child Health and Development Setagaya-ku Tokyo Japan
| | - Takeshi Takajo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Akinori Wada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Kazuki Horiuchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Kenichi Inaba
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Yoshinori Hanawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Naoki Shibuya
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Yoshikiyo Okada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Chie Kurihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Shin Nishii
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Akinori Mizoguchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Hideaki Hozumi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Chikako Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Shunsuke Komoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Shigeaki Nagao
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Junji Yamamoto
- Department of Pathology National Center for Child Health and Development Setagaya-ku Tokyo Japan
| | - Soichiro Miura
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan.,International University of Health and Welfare Graduate School Minato-ku Tokyo Japan
| | - Ryota Hokari
- Division of Gastroenterology and Hepatology, Department of Internal Medicine National Defense Medical College Tokorozawa-shi Saitama Japan
| | - Tananori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine Keio University School of Medicine Shinjuku-ku Tokyo Japan
| |
Collapse
|
88
|
Xu L, Wang W, Zhang X, Ke H, Qin Y, You L, Li W, Lu G, Chan WY, Leung PCK, Zhao S, Chen ZJ. Palmitic acid causes insulin resistance in granulosa cells via activation of JNK. J Mol Endocrinol 2019; 62:197-206. [PMID: 30913535 DOI: 10.1530/jme-18-0214] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/15/2022]
Abstract
Obesity is a worldwide health problem with rising incidence and results in reproductive difficulties. Elevated saturated free fatty acids (FFAs) in obesity can cause insulin resistance (IR) in peripheral tissues. The high intra-follicular saturated FFAs may also account for IR in ovarian granulosa cells (GCs). In the present study, we investigated the relationship between saturated FFAs and IR in GCs by the use of palmitic acid (PA). We demonstrated that the glucose uptake in cultured GCs and lactate accumulation in the culture medium were stimulated by insulin, but the effects of insulin were attenuated by PA treatment. Besides, insulin-induced phosphorylation of Akt was reduced by PA in a dose- and time-dependent manner. Furthermore, PA increased phosphorylation of JNK and JNK blockage rescued the phosphorylation of Akt which was downregulated by PA. These findings highlighted the negative effect of PA on GCs metabolism and may partially account for the obesity-related reproductive disorders.
Collapse
Affiliation(s)
- Lan Xu
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Wenting Wang
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- The Second Hospital of Shandong University, Jinan, China
| | - Xinyue Zhang
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Hanni Ke
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Yingying Qin
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Li You
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Weiping Li
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Gang Lu
- The CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wai-Yee Chan
- The CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Peter C K Leung
- Department of Obstetrics and Gynecology, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, Canada
| | - Shidou Zhao
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- The CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Shandong Provincial Hospital Affiliated to Shandong University, National Research Center for Assisted Reproductive Technology and Reproductive Genetics, The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
89
|
Badi RM, Mostafa DG, Khaleel EF, Satti HH. Resveratrol protects against hepatic insulin resistance in a rat's model of non-alcoholic fatty liver disease by down-regulation of GPAT-1 and DGAT2 expression and inhibition of PKC membranous translocation. Clin Exp Pharmacol Physiol 2019; 46:545-555. [PMID: 30773673 DOI: 10.1111/1440-1681.13074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/16/2019] [Accepted: 02/13/2019] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with hepatic insulin resistance (IR). Resveratrol (RES) a potent hypolipidemic dietary polyphenol has been identified for its ability to prevent hepatic steatosis and hepatic IR in high-fat diet (HFD)-fed murine models of NAFLD. In the present study, we have carried an in vivo animal experiment to identify a novel mechanism for RES protective action. Sub-chronic (45 days) RES pretreatment in 3 days HFD-fed adult Wistar rats prevented early hepatic IR through inhibiting PKC/JNK activation; decreasing p-IRS (Ser307 ) and increasing p-IRS(Tyr612 ), p-Akt(Ser473 ) and p-GSK3(Ser9 ). These effects of RES were associated with reduced expression of acyl-CoA:glycerol-sn-3-phosphate acyltransferase (GPAT-1) and diacylglycerol:acyl-CoA acyltransferase (DGAT2), two critical enzymes in the glycerol-3-phosphate pathway for de novo triglycerides synthesis. These data indicate that RES protects against NAFLD, initially, by inhibiting the early development of hepatic IR.
Collapse
Affiliation(s)
- Rehab M Badi
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Physiology, University of Khartoum, Khartoum, Sudan
| | - Dalia G Mostafa
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Medical Physiology, Assiut University, Assiut, Egypt
| | - Eman F Khaleel
- Department of Medical Physiology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Medical Physiology, Cairo University, Cairo, Egypt
| | - Huda H Satti
- Department of Pathology, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia.,Faculty of Medicine, Department of Pathology, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
90
|
Atawia RT, Bunch KL, Toque HA, Caldwell RB, Caldwell RW. Mechanisms of obesity-induced metabolic and vascular dysfunctions. FRONT BIOSCI-LANDMRK 2019; 24:890-934. [PMID: 30844720 PMCID: PMC6689231 DOI: 10.2741/4758] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has reached epidemic proportions and its prevalence is climbing. Obesity is characterized by hypertrophied adipocytes with a dysregulated adipokine secretion profile, increased recruitment of inflammatory cells, and impaired metabolic homeostasis that eventually results in the development of systemic insulin resistance, a phenotype of type 2 diabetes. Nitric oxide synthase (NOS) is an enzyme that converts L-arginine to nitric oxide (NO), which functions to maintain vascular and adipocyte homeostasis. Arginase is a ureohydrolase enzyme that competes with NOS for L-arginine. Arginase activity/expression is upregulated in obesity, which results in diminished bioavailability of NO, impairing both adipocyte and vascular endothelial cell function. Given the emerging role of NO in the regulation of adipocyte physiology and metabolic capacity, this review explores the interplay between arginase and NO, and their effect on the development of metabolic disorders, cardiovascular diseases, and mitochondrial dysfunction in obesity. A comprehensive understanding of the mechanisms involved in the development of obesity-induced metabolic and vascular dysfunction is necessary for the identification of more effective and tailored therapeutic avenues for their prevention and treatment.
Collapse
Affiliation(s)
- Reem T Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Katharine L Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology,and Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Robert W Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904,USA,
| |
Collapse
|
91
|
Baldassano S, Gasbjerg LS, Kizilkaya HS, Rosenkilde MM, Holst JJ, Hartmann B. Increased Body Weight and Fat Mass After Subchronic GIP Receptor Antagonist, but Not GLP-2 Receptor Antagonist, Administration in Rats. Front Endocrinol (Lausanne) 2019; 10:492. [PMID: 31447774 PMCID: PMC6691063 DOI: 10.3389/fendo.2019.00492] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/08/2019] [Indexed: 12/20/2022] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-2 (GLP-2) are hormones secreted from the enteroendocrine cells after a meal. They exert their actions through activation of G protein-coupled receptors (R), the GIPR and GLP-2R, respectively. Both have been reported to influence metabolism. The purpose of the study was to investigate the role of the hormones in the regulation of lipid and bone homeostasis by subchronic treatment with novel GIPR and GLP-2R antagonists. Rats were injected once daily with vehicle, GIPR, or GLP-2R antagonists for 3 weeks. Body weight, food intake, body composition, plasma lipoprotein lipase (LPL), adipokines, triglycerides and the marker of bone resorption carboxy-terminal collagen crosslinks (CTX), were examined. In rats, subchronic treatment with GIPR antagonist, rat GIP (3-30)NH2, did not modify food intake and bone resorption, but significantly increased body weight, body fat mass, triglycerides, LPL, and leptin levels compared with vehicle treated rats. Subchronic (Pro3)GIP (a partial GIPR agonist), GLP-2(11-33), and GLP-2(3-33) (GLP-2R antagonists) treatment did not affect any parameter. The present results would be consistent with a role for GIP, but not GLP-2, in the maintenance of lipid homeostasis in rats, while neither GIPR nor GLP-2R antagonism appeared to influence bone resorption in rats.
Collapse
Affiliation(s)
- Sara Baldassano
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Palermo, Italy
| | - Lærke Smidt Gasbjerg
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Bolette Hartmann
| |
Collapse
|
92
|
Metcalfe LK, Smith GC, Turner N. Defining lipid mediators of insulin resistance: controversies and challenges. J Mol Endocrinol 2019; 62:R65-R82. [PMID: 30068522 DOI: 10.1530/jme-18-0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/04/2018] [Accepted: 07/31/2018] [Indexed: 12/31/2022]
Abstract
Essential elements of all cells - lipids - play important roles in energy production, signalling and as structural components. Despite these critical functions, excessive availability and intracellular accumulation of lipid is now recognised as a major factor contributing to many human diseases, including obesity and diabetes. In the context of these metabolic disorders, ectopic deposition of lipid has been proposed to have deleterious effects on insulin action. While this relationship has been recognised for some time now, there is currently no unifying mechanism to explain how lipids precipitate the development of insulin resistance. This review summarises the evidence linking specific lipid molecules to the induction of insulin resistance, describing some of the current controversies and challenges for future studies in this field.
Collapse
Affiliation(s)
- Louise K Metcalfe
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, New South Wales, Australia
| | - Greg C Smith
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, New South Wales, Australia
| | - Nigel Turner
- Department of Pharmacology, School of Medical Sciences, UNSW Sydney, New South Wales, Australia
| |
Collapse
|
93
|
Petersen MC, Shulman GI. Mechanisms of Insulin Action and Insulin Resistance. Physiol Rev 2018; 98:2133-2223. [PMID: 30067154 PMCID: PMC6170977 DOI: 10.1152/physrev.00063.2017] [Citation(s) in RCA: 1701] [Impact Index Per Article: 243.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/22/2018] [Accepted: 03/24/2018] [Indexed: 12/15/2022] Open
Abstract
The 1921 discovery of insulin was a Big Bang from which a vast and expanding universe of research into insulin action and resistance has issued. In the intervening century, some discoveries have matured, coalescing into solid and fertile ground for clinical application; others remain incompletely investigated and scientifically controversial. Here, we attempt to synthesize this work to guide further mechanistic investigation and to inform the development of novel therapies for type 2 diabetes (T2D). The rational development of such therapies necessitates detailed knowledge of one of the key pathophysiological processes involved in T2D: insulin resistance. Understanding insulin resistance, in turn, requires knowledge of normal insulin action. In this review, both the physiology of insulin action and the pathophysiology of insulin resistance are described, focusing on three key insulin target tissues: skeletal muscle, liver, and white adipose tissue. We aim to develop an integrated physiological perspective, placing the intricate signaling effectors that carry out the cell-autonomous response to insulin in the context of the tissue-specific functions that generate the coordinated organismal response. First, in section II, the effectors and effects of direct, cell-autonomous insulin action in muscle, liver, and white adipose tissue are reviewed, beginning at the insulin receptor and working downstream. Section III considers the critical and underappreciated role of tissue crosstalk in whole body insulin action, especially the essential interaction between adipose lipolysis and hepatic gluconeogenesis. The pathophysiology of insulin resistance is then described in section IV. Special attention is given to which signaling pathways and functions become insulin resistant in the setting of chronic overnutrition, and an alternative explanation for the phenomenon of ‟selective hepatic insulin resistanceˮ is presented. Sections V, VI, and VII critically examine the evidence for and against several putative mediators of insulin resistance. Section V reviews work linking the bioactive lipids diacylglycerol, ceramide, and acylcarnitine to insulin resistance; section VI considers the impact of nutrient stresses in the endoplasmic reticulum and mitochondria on insulin resistance; and section VII discusses non-cell autonomous factors proposed to induce insulin resistance, including inflammatory mediators, branched-chain amino acids, adipokines, and hepatokines. Finally, in section VIII, we propose an integrated model of insulin resistance that links these mediators to final common pathways of metabolite-driven gluconeogenesis and ectopic lipid accumulation.
Collapse
Affiliation(s)
- Max C Petersen
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Howard Hughes Medical Institute, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
94
|
Pi-Sunyer X. Changes in body composition and metabolic disease risk. Eur J Clin Nutr 2018; 73:231-235. [PMID: 30275524 DOI: 10.1038/s41430-018-0320-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 11/09/2022]
Abstract
As individuals gain weight, they increase the amount of fat that they accrue on their body. This causes adipocytes to enlarge and increases not only subcutaneous fat but also deposits fat in other vulnerable areas of the body. This ectopic fat is deposited in the intra-abdominal visceral fat depot, in muscle, in the liver and in the beta cells. Fat in these locations initiates a dysfunctional state in these insulin-sensitive tissues leading to insulin resistance, the appearance of the Metabolic Syndrome, and an increased risk of developing both type 2 diabetes and cardiovascular disease. A loss of weight and with it a loss of fat decreases this risk.
Collapse
Affiliation(s)
- Xavier Pi-Sunyer
- Columbia University College of Physicians and Surgeons and Columbia Institute of Human Nutrition, New York, NY, USA.
| |
Collapse
|
95
|
Bigagli E, Cinci L, Niccolai A, Biondi N, Rodolfi L, D'Ottavio M, D'Ambrosio M, Lodovici M, Tredici MR, Luceri C. Preliminary data on the dietary safety, tolerability and effects on lipid metabolism of the marine microalga Tisochrysis lutea. ALGAL RES 2018. [DOI: 10.1016/j.algal.2018.08.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
96
|
Su Y, Shen X, Chen J, Isales CM, Zhao J, Shi XM. Differentially expressed genes in PPARγ-deficient MSCs. Mol Cell Endocrinol 2018; 471:97-104. [PMID: 28774780 PMCID: PMC5792374 DOI: 10.1016/j.mce.2017.07.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 07/26/2017] [Accepted: 07/28/2017] [Indexed: 12/28/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a key regulator of adipogenesis. It is also a central player in energy metabolism, inflammation and immunity. As an important nuclear transcription factor, PPARγ can regulate the expression and function of genes or biological processes directly or indirectly via association with other factors and thus modulate their activities. To better understand the impact of PPARγ on the global gene expression profile, we evaluated the bioinformatic data, which revealed the changes that occurred in genes and their pathways in the absence of PPARγ. In brief, we performed RNA deep sequencing (RNA-Seq) analysis using RNA samples isolated from multipotent mesenchymal stromal cells (MSCs) of PPARγ knockout and wild type control mice. The RNA-Seq data sets were then subjected to bioinformatic analyses from various angles to better reveal the breadth of PPARγ function in different biological processes. Our results reveal novel genes and networks modulated by PPARγ and provides new insights into our understanding of the physiologic and pathophysiologic role this nuclear receptor plays in health and disease.
Collapse
Affiliation(s)
- Yun Su
- Department of Neuroscience & Regenerative Medicine, USA
| | - Xiaona Shen
- Department of Mathematics, Logistical Engineering University, Chongqing, China
| | - Jie Chen
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, GA, USA
| | - Carlos M Isales
- Department of Neuroscience & Regenerative Medicine, USA; Orthopaedic Surgery, Augusta University, Augusta, GA, USA
| | - Jing Zhao
- Department of Mathematics, Logistical Engineering University, Chongqing, China.
| | - Xing-Ming Shi
- Department of Neuroscience & Regenerative Medicine, USA; Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
97
|
Zhao W, Feng X, Liu B, Xian J, Zhang N. Er-Miao-Fang Extracts Inhibits Adipose Lipolysis and Reduces Hepatic Gluconeogenesis via Suppression of Inflammation. Front Physiol 2018; 9:1041. [PMID: 30154727 PMCID: PMC6102449 DOI: 10.3389/fphys.2018.01041] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 07/12/2018] [Indexed: 01/07/2023] Open
Abstract
High-fat-diet (HFD) feeding induces adipose dysfunction. This study aims to explore whether the Traditional Chinese Medical prescription Er-Miao-Fang could ameliorate adipose dysfunction and prevent hepatic glucose output. Short-term HFD feeding induced adipose lipolysis accompanied with enhanced hepatic glucose output in mice. Adipose lipolysis is initiated by cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling. Oral administration Er-Miao-Fang inhibited inflammation in adipose tissue by dephosphorylation of JNK and reducing TNF-α and IL-1β production, and thus preserved phosphodiesterase 3B (PDE3B) induction, contributing to preventing cAMP accumulation. As a result, from suppression of PKA activation, Er-Miao-Fang reduced fatty acids and glycerol release from adipose tissue due to the inhibition hormone-sensitive lipase (HSL). By blocking the traffic of fatty acids and inflammatory mediators from adipose tissue to the liver, Er-Miao-Fang attenuated hepatic cAMP/PKA signaling by protecting phosphodiesterase 4B (PDE4B) induction from inflammatory insult, and thereby reduced hepatic glucose production by suppression of hepatic glucagon response in HFD-fed mice. In conclusion, Er-Miao-Fang prevented adipose lipolysis by suppression of inflammation, contributing to reducing excessive hepatic glucose output. These findings present a new view of regulating gluconeogenesis and provide the guiding significance for the regulation of multi-link targets with Traditional Chinese Medicine.
Collapse
Affiliation(s)
- Wenjun Zhao
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xin Feng
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baolin Liu
- Clinical Metabolomics Centre, China Pharmaceutical University, Nanjing, China
| | - Jiechen Xian
- Engineering Research Center of Modern Preparation Technology of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
98
|
Sendi H, Mead I, Wan M, Mehrab-Mohseni M, Koch K, Atala A, Bonkovsky HL, Bishop CE. miR-122 inhibition in a human liver organoid model leads to liver inflammation, necrosis, steatofibrosis and dysregulated insulin signaling. PLoS One 2018; 13:e0200847. [PMID: 30024933 PMCID: PMC6053181 DOI: 10.1371/journal.pone.0200847] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/03/2018] [Indexed: 12/12/2022] Open
Abstract
To investigate the role of miR-122 in the development and regression of non-alcoholic fatty liver disease (NAFLD) in vitro, we used multicellular 3D human liver organoids developed in our laboratory. These organoids consist of primary human hepatocytes, Kupffer cells, quiescent stellate cells and liver sinusoidal endothelial cells. They remain viable and functional for 4 weeks expressing typical markers of liver function such as synthesis of albumin, urea, and alpha-1 p450 drug metabolism. Before mixing, hepatic cells were transduced with lentivirus to inhibit miR122 expression (ABM, CA). Immediately after the organoids were fully formed (day 4) or after 1 or 2 weeks of additional incubation (days 11 or 18), the organoids were analyzed using fluorescent live/dead staining and ATP production; total RNA was extracted for qPCR gene expression profiling. Our results show that miR-122 inhibition in liver organoids leads to inflammation, necrosis, steatosis and fibrosis. This was associated with increase in inflammatory cytokines (IL6, TNF), chemokines (CCL2, CCL3) and increase in a subset of Matrix Metaloproteinases (MMP8, MMP9). An altered expression of key genes in lipid metabolism (i.e LPL, LDLR) and insulin signaling (i.e GLUT4, IRS1) was also identified. Conclusion: Our results highlight the role of miR-122 inhibition in liver inflammation, steatofibrosis and dysregulation of insulin signaling. Patients with NAFLD are known to have altered levels of miR-122, therefore we suggest that miR-122 mimics could play a useful role in reversing liver steatofibrosis and insulin resistance seen in patients with NAFLD.
Collapse
Affiliation(s)
- Hossein Sendi
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
- * E-mail:
| | - Ivy Mead
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Meimei Wan
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Marjan Mehrab-Mohseni
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Kenneth Koch
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Anthony Atala
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| | - Herbert L. Bonkovsky
- The Laboratory for Liver Diseases and Metabolic Disorders, Section on Gastroenterology, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Colin E. Bishop
- Wake Forest Institute of Regenerative Medicine, Winston-Salem, NC, United States of America
| |
Collapse
|
99
|
Tao R, Wang C, Stöhr O, Qiu W, Hu Y, Miao J, Dong XC, Leng S, Stefater M, Stylopoulos N, Lin L, Copps KD, White MF. Inactivating hepatic follistatin alleviates hyperglycemia. Nat Med 2018; 24:1058-1069. [PMID: 29867232 PMCID: PMC6039237 DOI: 10.1038/s41591-018-0048-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 04/10/2018] [Indexed: 12/16/2022]
Abstract
Unsuppressed hepatic glucose production (HGP) contributes substantially to glucose intolerance and diabetes, which can be modeled by the genetic inactivation of hepatic insulin receptor substrate 1 (Irs1) and Irs2 (LDKO mice). We previously showed that glucose intolerance in LDKO mice is resolved by hepatic inactivation of the transcription factor FoxO1 (that is, LTKO mice)-even though the liver remains insensitive to insulin. Here, we report that insulin sensitivity in the white adipose tissue of LDKO mice is also impaired but is restored in LTKO mice in conjunction with normal suppression of HGP by insulin. To establish the mechanism by which white adipose tissue insulin signaling and HGP was regulated by hepatic FoxO1, we identified putative hepatokines-including excess follistatin (Fst)-that were dysregulated in LDKO mice but normalized in LTKO mice. Knockdown of hepatic Fst in the LDKO mouse liver restored glucose tolerance, white adipose tissue insulin signaling and the suppression of HGP by insulin; however, the expression of Fst in the liver of healthy LTKO mice had the opposite effect. Of potential clinical significance, knockdown of Fst also improved glucose tolerance in high-fat-fed obese mice, and the level of serum Fst was reduced in parallel with glycated hemoglobin in obese individuals with diabetes who underwent therapeutic gastric bypass surgery. We conclude that Fst is a pathological hepatokine that might be targeted for diabetes therapy during hepatic insulin resistance.
Collapse
Affiliation(s)
- Rongya Tao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caixia Wang
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oliver Stöhr
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Qiu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yue Hu
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - X Charlie Dong
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sining Leng
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Margaret Stefater
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas Stylopoulos
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lin Lin
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kyle D Copps
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Morris F White
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
100
|
Morales-Prieto N, Ruiz-Laguna J, Sheehan D, Abril N. Transcriptome signatures of p,p´-DDE-induced liver damage in Mus spretus mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:150-167. [PMID: 29554563 DOI: 10.1016/j.envpol.2018.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/19/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023]
Abstract
The use of DDT (1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane) in some countries, although regulated, is contributing to an increased worldwide risk of exposure to this organochlorine pesticide or its derivative p,p'-DDE [1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene]. Many studies have associated p,p'-DDE exposure to type 2 diabetes, obesity and alterations of the reproductive system, but their molecular mechanisms of toxicity remain poorly understood. We have addressed this issue by using commercial microarrays based on probes for the entire Mus musculus genome to determine the hepatic transcriptional signatures of p,p'-DDE in the phylogenetically close mouse species Mus spretus. High-stringency hybridization conditions and analysis assured reliable results, which were also verified, in part, by qRT-PCR, immunoblotting and/or enzymatic activity. Our data linked 198 deregulated genes to mitochondrial dysfunction and perturbations of central signaling pathways (kinases, lipids, and retinoic acid) leading to enhanced lipogenesis and aerobic glycolysis, inflammation, cell proliferation and testosterone catabolism and excretion. Alterations of transcript levels of genes encoding enzymes involved in testosterone catabolism and excretion would explain the relationships established between p,p´-DDE exposure and reproductive disorders, obesity and diabetes. Further studies will help to fully understand the molecular basis of p,p´-DDE molecular toxicity in liver and reproductive organs, to identify effective exposure biomarkers and perhaps to design efficient p,p'-DDE exposure counteractive strategies.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Julia Ruiz-Laguna
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - David Sheehan
- College of Arts and Science, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| |
Collapse
|