101
|
Gottlieb A, Leven AS, Sowa JP, Borucki K, Link A, Yilmaz E, Aygen S, Canbay A, Porsch-Özcürümez M. Lipoprotein and Metabolic Profiles Indicate Similar Cardiovascular Risk of Liver Steatosis and NASH. Digestion 2020; 102:671-681. [PMID: 33080603 DOI: 10.1159/000510600] [Citation(s) in RCA: 5] [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] [Received: 05/05/2020] [Accepted: 07/29/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Nonalcoholic fatty liver disease (NAFLD) affects about 25% of the global population, with no reliable noninvasive tests to diagnose nonalcoholic steatohepatitis (NASH) and to differentiate between NASH and nonalcoholic fatty liver (NAFL) (steatosis alone). It is unclear if NAFL and NASH differ in cardiovascular risk for patients. Here, we compared obese NAFLD patients with a healthy cohort to test whether cholesterol compounds could represent potential noninvasive markers and to estimate associated risks. METHOD Serum samples of 46 patients with histologically confirmed NAFLD (17 NAFL, 29 NASH) who underwent bariatric surgery were compared to 32 (9 males, 21 females) healthy controls (HCs). We analyzed epidemiological data, liver enzymes, cholesterol and lipid profile, and amino acids. The latter were analyzed by nuclear magnetic resonance spectroscopy. RESULTS Total serum and high-density lipoprotein (HDL) cholesterol were significantly lower in the NAFLD group than in HCs, with a stronger reduction in NASH. Similar observations were made for sub-specification of HDL-p, HDL-s, SHDL-p, and LHDL-p cholesterols. Low-density lipoprotein (LDL)-s and LLDL-p cholesterol were significantly reduced in NAFLD groups. Interestingly, SLDL-p cholesterol was significantly higher in the NAFL group with a stronger elevation in NASH than in HCs. The amino acids alanine, leucin, and isoleucine were significantly higher in the NAFL and NASH groups than in HCs. CONCLUSION We show in this study that cholesterol profiles, apolipoproteins, and amino acids could function as a potential noninvasive test to screen for NAFLD or even NASH in larger populations. However, few differences in cholesterol profiles were identified between the NAFL and NASH groups, indicating similar cardiovascular risk profiles.
Collapse
Affiliation(s)
- Aline Gottlieb
- Department of Physiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anna-Sophia Leven
- Department for General- and Visceral Surgery, Alfried Krupp Hospital, Essen, Germany
| | - Jan-Peter Sowa
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, University Bochum, Bochum, Germany
| | - Katrin Borucki
- Institute for Clinical Chemistry and Pathobiochemistry, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology, and Infectious Diseases, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | | | | - Ali Canbay
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, University Bochum, Bochum, Germany,
| | - Mustafa Porsch-Özcürümez
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, University Bochum, Bochum, Germany
| |
Collapse
|
102
|
Li X, Sun D, Zhou T, Ma H, Heianza Y, Liang Z, Bray GA, Sacks FM, Qi L. Changes of Branched-Chain Amino Acids and Ectopic Fat in Response to Weight-loss Diets: the POUNDS Lost Trial. J Clin Endocrinol Metab 2020; 105:e3747-e3756. [PMID: 32561903 PMCID: PMC7465554 DOI: 10.1210/clinem/dgaa377] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/11/2020] [Indexed: 12/25/2022]
Abstract
CONTEXT Recent evidence has related circulating branch-chained amino acids (BCAAs) to ectopic fat distribution. OBJECTIVE To investigate the associations of changes in plasma BCAAs induced by weight-loss diet interventions with hepatic fat and abdominal fat, and potential modification by different diets. DESIGN, SETTING, AND PARTICIPANTS The current study included 184 participants from the 2-year Preventing Overweight and Using Novel Dietary Strategies (POUNDS Lost) trial with repeated measurements on plasma BCAAs, hepatic fat, and abdominal fat over 2 years. MAIN OUTCOME MEASURES Repeated measurements of hepatic fat, abdominal fat distribution, including visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), and total adipose tissue (TAT). RESULTS Over 2 years, a decrease in total plasma BCAAs was significantly associated with improvement in hepatic density (a marker for hepatic fat; P = 0.02) and reductions in abdominal fat, including VAT, SAT, and TAT (all P < 0.05) in the main analyses. Additionally, we observed that decreases in BCAAs were associated with decreased insulin, homeostasis model assessment of insulin resistance, and triglycerides, independent of weight loss (all P < 0.05). Moreover, we found that dietary protein intake significantly modified the relation between changes in total plasma BCAAs and hepatic density at 6 months (Pinteraction = 0.01). Participants with a larger decrease in total BCAAs showed a greater increase in hepatic density when consuming a high-protein diet, compared with those with a smaller decrease or increase in total BCAAs. CONCLUSIONS Our findings indicate that weight-loss diet-induced decrease in plasma BCAAs is associated with reductions of hepatic and abdominal fat. In addition, dietary protein intake may modify these associations.
Collapse
Affiliation(s)
- Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Dianjianyi Sun
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Tao Zhou
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Hao Ma
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
| | - Zhaoxia Liang
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
- Department of Obstetrical, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - George A Bray
- Pennington Biomedical Research Center of the Louisiana State University System, Baton Rouge, Louisiana
| | - Frank M Sacks
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
103
|
Hamill MJ, Afeyan R, Chakravarthy MV, Tramontin T. Endogenous Metabolic Modulators: Emerging Therapeutic Potential of Amino Acids. iScience 2020; 23:101628. [PMID: 33103071 PMCID: PMC7569218 DOI: 10.1016/j.isci.2020.101628] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multifactorial disease pathophysiology is complex and incompletely addressed by existing targeted pharmacotherapies. Amino acids (AAs) and related metabolites and precursors are a class of endogenous metabolic modulators (EMMs) that have diverse biological functions and, thus, have been explored for decades as potential multifactorial disease treatments. Here, we review the literature on this class of EMMs in disease treatment, with a focus on the emerging clinical studies on AAs and related metabolites and precursors as single- and combination-agents targeted to a single biology. These clinical research insights, in addition to increasing understanding of disease metabolic profiles and combinatorial therapeutic design principles, highlight an opportunity to develop EMM compositions with AAs and related metabolites and precursors to target multifactorial disease biology. EMM compositions are uniquely designed to enable a comprehensive approach, with potential to simultaneously and safely target pathways underlying multifactorial diseases and to regulate biological processes that promote overall health.
Collapse
|
104
|
Della Torre S. Non-alcoholic Fatty Liver Disease as a Canonical Example of Metabolic Inflammatory-Based Liver Disease Showing a Sex-Specific Prevalence: Relevance of Estrogen Signaling. Front Endocrinol (Lausanne) 2020; 11:572490. [PMID: 33071979 PMCID: PMC7531579 DOI: 10.3389/fendo.2020.572490] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
There is extensive evidence supporting the interplay between metabolism and immune response, that have evolved in close relationship, sharing regulatory molecules and signaling systems, to support biological functions. Nowadays, the disruption of this interaction in the context of obesity and overnutrition underlies the increasing incidence of many inflammatory-based metabolic diseases, even in a sex-specific fashion. During evolution, the interplay between metabolism and reproduction has reached a degree of complexity particularly high in female mammals, likely to ensure reproduction only under favorable conditions. Several factors may account for differences in the incidence and progression of inflammatory-based metabolic diseases between females and males, thus contributing to age-related disease development and difference in life expectancy between the two sexes. Among these factors, estrogens, acting mainly through Estrogen Receptors (ERs), have been reported to regulate several metabolic pathways and inflammatory processes particularly in the liver, the metabolic organ showing the highest degree of sexual dimorphism. This review aims to investigate on the interaction between metabolism and inflammation in the liver, focusing on the relevance of estrogen signaling in counteracting the development and progression of non-alcoholic fatty liver disease (NAFLD), a canonical example of metabolic inflammatory-based liver disease showing a sex-specific prevalence. Understanding the role of estrogens/ERs in the regulation of hepatic metabolism and inflammation may provide the basis for the development of sex-specific therapeutic strategies for the management of such an inflammatory-based metabolic disease and its cardio-metabolic consequences.
Collapse
Affiliation(s)
- Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
105
|
Perakakis N, Joshi A, Peradze N, Stefanakis K, Li G, Feigh M, Veidal SS, Rosen G, Fleming M, Mantzoros CS. The Selective Peroxisome Proliferator-Activated Receptor Gamma Modulator CHS-131 Improves Liver Histopathology and Metabolism in a Mouse Model of Obesity and Nonalcoholic Steatohepatitis. Hepatol Commun 2020; 4:1302-1315. [PMID: 32923834 PMCID: PMC7471426 DOI: 10.1002/hep4.1558] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/18/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
CHS-131 is a selective peroxisome proliferator-activated receptor gamma modulator with antidiabetic effects and less fluid retention and weight gain compared to thiazolidinediones in phase II clinical trials. We investigated the effects of CHS-131 on metabolic parameters and liver histopathology in a diet-induced obese (DIO) and biopsy-confirmed mouse model of nonalcoholic steatohepatitis (NASH). Male C57BL/6JRj mice were fed the amylin liver NASH diet (40% fat with trans-fat, 20% fructose, and 2% cholesterol). After 36 weeks, only animals with biopsy-confirmed steatosis and fibrosis were included and stratified into treatment groups (n = 12-13) to receive for the next 12 weeks (1) low-dose CHS-131 (10 mg/kg), (2) high-dose CHS-131 (30 mg/kg), or (3) vehicle. Metabolic parameters, liver pathology, metabolomics/lipidomics, markers of liver function and liver, and subcutaneous and visceral adipose tissue gene expression profiles were assessed. CHS-131 did not affect body weight, fat mass, lean mass, water mass, or food intake in DIO-NASH mice with fibrosis. CHS-131 improved fasting insulin levels and insulin sensitivity as assessed by the intraperitoneal insulin tolerance test. CHS-131 improved total plasma cholesterol, triglycerides, alanine aminotransferase, and aspartate aminotransferase and increased plasma adiponectin levels. CHS-131 (high dose) improved liver histology and markers of hepatic fibrosis. DIO-NASH mice treated with CHS-131 demonstrated a hepatic shift to diacylglycerols and triacylglycerols with a lower number of carbons, increased expression of genes stimulating fatty acid oxidation and browning, and decreased expression of genes promoting fatty acid synthesis, triglyceride synthesis, and inflammation in adipose tissue. Conclusion: CHS-131 improves liver histology in a DIO and biopsy-confirmed mouse model of NASH by altering the hepatic lipidome, reducing insulin resistance, and improving lipid metabolism and inflammation in adipose tissue.
Collapse
Affiliation(s)
- Nikolaos Perakakis
- Department of Internal MedicineBoston VA Healthcare SystemBostonMA
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
| | - Aditya Joshi
- Department of Internal MedicineBoston VA Healthcare SystemBostonMA
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
| | - Natia Peradze
- Department of Internal MedicineBoston VA Healthcare SystemBostonMA
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
| | - Konstantinos Stefanakis
- Department of Internal MedicineBoston VA Healthcare SystemBostonMA
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
| | | | | | | | | | | | - Christos S. Mantzoros
- Department of Internal MedicineBoston VA Healthcare SystemBostonMA
- Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMA
| |
Collapse
|
106
|
Fougerat A, Montagner A, Loiseau N, Guillou H, Wahli W. Peroxisome Proliferator-Activated Receptors and Their Novel Ligands as Candidates for the Treatment of Non-Alcoholic Fatty Liver Disease. Cells 2020; 9:E1638. [PMID: 32650421 PMCID: PMC7408116 DOI: 10.3390/cells9071638] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, frequently associated with obesity and type 2 diabetes. Steatosis is the initial stage of the disease, which is characterized by lipid accumulation in hepatocytes, which can progress to non-alcoholic steatohepatitis (NASH) with inflammation and various levels of fibrosis that further increase the risk of developing cirrhosis and hepatocellular carcinoma. The pathogenesis of NAFLD is influenced by interactions between genetic and environmental factors and involves several biological processes in multiple organs. No effective therapy is currently available for the treatment of NAFLD. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that regulate many functions that are disturbed in NAFLD, including glucose and lipid metabolism, as well as inflammation. Thus, they represent relevant clinical targets for NAFLD. In this review, we describe the determinants and mechanisms underlying the pathogenesis of NAFLD, its progression and complications, as well as the current therapeutic strategies that are employed. We also focus on the complementary and distinct roles of PPAR isotypes in many biological processes and on the effects of first-generation PPAR agonists. Finally, we review novel and safe PPAR agonists with improved efficacy and their potential use in the treatment of NAFLD.
Collapse
Affiliation(s)
- Anne Fougerat
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Alexandra Montagner
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Institut National de la Santé et de la Recherche Médicale (Inserm), Institute of Metabolic and Cardiovascular Diseases, UMR1048 Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases, University of Toulouse, UMR1048 Toulouse, France
| | - Nicolas Loiseau
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Hervé Guillou
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Walter Wahli
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
107
|
Grzych G, Vonghia L, Bout MA, Weyler J, Verrijken A, Dirinck E, Chevalier Curt MJ, Van Gaal L, Paumelle R, Francque S, Tailleux A, Haas JT, Staels B. Plasma BCAA Changes in Patients With NAFLD Are Sex Dependent. J Clin Endocrinol Metab 2020; 105:5818376. [PMID: 32271385 DOI: 10.1210/clinem/dgaa175] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 04/08/2020] [Indexed: 12/15/2022]
Abstract
CONTEXT Plasma branched chain amino acid (BCAA) concentrations correlate positively with body mass index (BMI), measures of insulin resistance (IR), and severity of nonalcoholic fatty liver disease (NAFLD). Moreover, plasma BCAA concentrations also differ between the sexes, which display different susceptibilities to cardio-metabolic diseases. OBJECTIVE Assess whether plasma BCAA concentrations associate with NAFLD severity independently of BMI, IR, and sex. PATIENTS Patients visiting the obesity clinic of the Antwerp University Hospital were consecutively recruited from 2006 to 2014. DESIGN AND SETTING A cross-sectional study cohort of 112 obese patients (59 women and 53 men) was divided into 4 groups according to NAFLD severity. Groups were matched for sex, age, BMI, homeostatic model assessment of IR, and hemoglobin A1c. MAIN OUTCOME MEASURES Fasting plasma BCAA concentrations were measured by tandem mass spectrometry using the aTRAQ™ method. RESULTS In the study cohort, a modest positive correlation was observed between plasma BCAA concentrations and NAFLD severity, as well as a strong effect of sex on plasma BCAA levels. Subgroup analysis by sex revealed that while plasma BCAA concentrations increased with severity of NAFLD in women, they tended to decrease in men. Additionally, only women displayed significantly increased plasma BCAAs with increasing fibrosis. CONCLUSION Plasma BCAA concentrations display sex-dimorphic changes with increasing severity of NAFLD, independently of BMI, IR, and age. Additionally, plasma BCAA are associated with significant fibrosis in women, but not in men. These results highlight the importance of a careful consideration of sex as a major confounding factor in cross-sectional studies of NAFLD.
Collapse
Affiliation(s)
- Guillaume Grzych
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
- CHU Lille, Service d'Hormonologie, Métabolisme, Nutrition, Oncologie, Lille, France
| | - Luisa Vonghia
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem/Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk/Antwerp, Belgium
| | - Marie-Adélaïde Bout
- CHU Lille, Service d'Hormonologie, Métabolisme, Nutrition, Oncologie, Lille, France
| | - Jonas Weyler
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem/Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk/Antwerp, Belgium
| | - An Verrijken
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk/Antwerp, Belgium
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem/Antwerp, Belgium
| | - Eveline Dirinck
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk/Antwerp, Belgium
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem/Antwerp, Belgium
| | | | - Luc Van Gaal
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk/Antwerp, Belgium
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Edegem/Antwerp, Belgium
| | - Réjane Paumelle
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem/Antwerp, Belgium
- Laboratory of Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk/Antwerp, Belgium
| | - Anne Tailleux
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
- CHU Lille, Service d'Hormonologie, Métabolisme, Nutrition, Oncologie, Lille, France
| |
Collapse
|
108
|
Changed Amino Acids in NAFLD and Liver Fibrosis: A Large Cross-Sectional Study without Influence of Insulin Resistance. Nutrients 2020; 12:nu12051450. [PMID: 32429590 PMCID: PMC7284573 DOI: 10.3390/nu12051450] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/06/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Altered amino acid levels have been found in nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). However, it is not clear whether this alteration is due to altered hepatic metabolism or insulin resistance. The aim of this study was to clarify the association among amino acid levels, fatty liver, and liver fibrosis while eliminating the influence of insulin resistance. NAFLD and liver fibrosis were diagnosed using transient elastography and subjects were divided into three groups: normal, NAFLD, and liver fibrosis. To exclude the influence of insulin resistance, the subjects were matched using the homeostasis model assessment of insulin resistance (HOMA-IR). The amino acid serum levels were compared among the groups. Of 731 enrolled subjects, 251 and 33 were diagnosed with NAFLD and liver fibrosis. Although significant differences were observed among the groups in the serum levels of most amino acids, all but those of glutamate and glycine disappeared after matching for HOMA-IR. The multivariate logistic regression revealed that glutamate, glycine, and HOMA-IR were independent risk factors for liver fibrosis. The altered serum levels of most amino acids were associated with insulin resistance, while the increase in glutamate and the decrease in glycine levels were strongly associated not only with insulin resistance, but also with altered liver metabolism in patients with liver fibrosis.
Collapse
|
109
|
Abstract
Coordinated changes in energy metabolism develop to support gestation and lactation in the periparturient dairy cow. Maternal physiology involves the partitioning of nutrients (i.e. glucose, amino acids and fatty acids (FA)) for fetal growth and milk synthesis. However, the inability of the dairy cow to successfully adapt to a productive lactation may trigger metabolic stress characterized by uncontrolled adipose tissue lipolysis and reduced insulin sensitivity. A consequence is lipotoxicity and hepatic triglyceride deposition that favors the development of fatty liver disease (FLD) and ketosis. This review describes contemporary perspectives pertaining to FA surfeit and complex lipid metabolism in the transition dairy cow. The role of saturated and unsaturated FA as bioactive signaling molecules capable of modulating insulin secretion and sensitivity is explored. Moreover, the metabolic fate of FA as influenced by mitochondrial function is considered. This includes the influence of inadequate mitochondrial oxidation on acylcarnitine status and the use of FA for lipid mediator synthesis. Lipid mediators, including the sphingolipid ceramide and diacylglycerol, are evaluated considering their established ability to inhibit insulin signaling and glucose transport in non-ruminant diabetics. The mechanisms of FLD in the transition cow are revisited with attention centered on glycerophospholipid phosphatidylcholine and triglyceride secretion. The relationship between oxidative stress and oxylipids within the context of insulin antagonism, hepatic steatosis and inflammation is also reviewed. Lastly, peripartal hormonal involvement or lack thereof of adipokines (i.e. leptin, adiponectin) and hepatokines (i.e. fibroblast growth factor-21) is described. Similarities and differences in ruminant and non-ruminant physiology are routinely showcased. Unraveling the lipidome of the dairy cow has generated breakthroughs in our understanding of periparturient lipid biology. Therapeutic approaches that target FA and complex lipid metabolism holds promise to enhance cow health, well-being and productive lifespan.
Collapse
|
110
|
Lonardo A, Suzuki A. Sexual Dimorphism of NAFLD in Adults. Focus on Clinical Aspects and Implications for Practice and Translational Research. J Clin Med 2020; 9:1278. [PMID: 32354182 PMCID: PMC7288212 DOI: 10.3390/jcm9051278] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) embraces the clinico-pathological consequences of hepatic lipotoxicity and is a major public health problem globally. Sexual dimorphism is a definite feature of most human diseases but, under this aspect, NAFLD lags behind other medical fields. Here, we aim at summarizing and critically discussing the most prominent sex differences and gaps in NAFLD in humans, with emphasis on those aspects which are relevant for clinical practice and translational research. Sexual dimorphism of NAFLD is covered with references to the following areas: disease prevalence and risk factors, pathophysiology, comorbidities, natural course and complications. Finally, we also discuss selected gender differences and whether sex-specific lifestyle changes should be adopted to contrast NAFLD in men and women.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Operating Unit Metabolic Syndrome, Azienda Ospedaliero-Universitaria di Modena, Ospedale Civile di Baggiovara, 41126 Baggiovara MO, Italy
| | - Ayako Suzuki
- Division of Gastroenterology, Durham VA Medical Center and Duke University Medical Center, Durham, NC 27705, USA;
| |
Collapse
|
111
|
Distinguishing NASH Histological Severity Using a Multiplatform Metabolomics Approach. Metabolites 2020; 10:metabo10040168. [PMID: 32344559 PMCID: PMC7240949 DOI: 10.3390/metabo10040168] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 12/31/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is categorized based on histological severity into nonalcoholic fatty liver (NAFL) or nonalcoholic steatohepatitis (NASH). We used a multiplatform metabolomics approach to identify metabolite markers and metabolic pathways that distinguish NAFL from early NASH and advanced NASH. We analyzed fasting serum samples from 57 prospectively-recruited patients with histologically-proven NAFLD, including 12 with NAFL, 31 with early NASH and 14 with advanced NASH. Metabolite profiling was performed using a combination of liquid chromatography-mass spectrometry (LC-MS) and nuclear magnetic resonance (NMR) spectroscopy analyzed with multivariate statistical and pathway analysis tools. We targeted 237 metabolites of which 158 were quantified. Multivariate analysis uncovered metabolite profile clusters for patients with NAFL, early NASH, and advanced NASH. Also, multiple individual metabolites were associated with histological severity, most notably spermidine which was more than 2-fold lower in advanced fibrosis vs. early fibrosis, in advanced NASH vs. NAFL and in advanced NASH vs. early NASH, suggesting that spermidine exercises a protective effect against development of fibrosing NASH. Furthermore, the results also showed metabolic pathway perturbations between early-NASH and advanced-NASH. In conclusion, using a combination of two reliable analytical platforms (LC-MS and NMR spectroscopy) we identified individual metabolites, metabolite clusters and metabolic pathways that were significantly different between NAFL, early-NASH, and advanced-NASH. These differences provide mechanistic insights as well as potentially important metabolic biomarker candidates that may noninvasively distinguish patients with NAFL, early-NASH, and advanced-NASH. The associations of spermidine levels with less advanced histology merit further assessment of the potential protective effects of spermidine in NAFLD.
Collapse
|
112
|
Felmlee MA, Jones RS, Rodriguez-Cruz V, Follman KE, Morris ME. Monocarboxylate Transporters (SLC16): Function, Regulation, and Role in Health and Disease. Pharmacol Rev 2020; 72:466-485. [PMID: 32144120 PMCID: PMC7062045 DOI: 10.1124/pr.119.018762] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The solute carrier family 16 (SLC16) is comprised of 14 members of the monocarboxylate transporter (MCT) family that play an essential role in the transport of important cell nutrients and for cellular metabolism and pH regulation. MCTs 1-4 have been extensively studied and are involved in the proton-dependent transport of L-lactate, pyruvate, short-chain fatty acids, and monocarboxylate drugs in a wide variety of tissues. MCTs 1 and 4 are overexpressed in a number of cancers, and current investigations have focused on transporter inhibition as a novel therapeutic strategy in cancers. MCT1 has also been used in strategies aimed at enhancing drug absorption due to its high expression in the intestine. Other MCT isoforms are less well characterized, but ongoing studies indicate that MCT6 transports xenobiotics such as bumetanide, nateglinide, and probenecid, whereas MCT7 has been characterized as a transporter of ketone bodies. MCT8 and MCT10 transport thyroid hormones, and recently, MCT9 has been characterized as a carnitine efflux transporter and MCT12 as a creatine transporter. Expressed at the blood brain barrier, MCT8 mutations have been associated with an X-linked intellectual disability, known as Allan-Herndon-Dudley syndrome. Many MCT isoforms are associated with hormone, lipid, and glucose homeostasis, and recent research has focused on their potential roles in disease, with MCTs representing promising novel therapeutic targets. This review will provide a summary of the current literature focusing on the characterization, function, and regulation of the MCT family isoforms and on their roles in drug disposition and in health and disease. SIGNIFICANCE STATEMENT: The 14-member solute carrier family 16 of monocarboxylate transporters (MCTs) plays a fundamental role in maintaining intracellular concentrations of a broad range of important endogenous molecules in health and disease. MCTs 1, 2, and 4 (L-lactate transporters) are overexpressed in cancers and represent a novel therapeutic target in cancer. Recent studies have highlighted the importance of MCTs in glucose, lipid, and hormone homeostasis, including MCT8 in thyroid hormone brain uptake, MCT12 in carnitine transport, and MCT11 in type 2 diabetes.
Collapse
Affiliation(s)
- Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Robert S Jones
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Kristin E Follman
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| | - Marilyn E Morris
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, California (M.A.F.); Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York (R.S.J., V.R.-C., M.E.M.); and Certara Strategic Consulting, Certara USA, Princeton, New Jersey (K.E.F.)
| |
Collapse
|
113
|
Tayyab Raza Naqvi S, Rasheed T, Naeem Ashiq M, Najam ul Haq M, Majeed S, Fatima B, Nawaz R, Hussain D, Shafi S. Fabrication of iron modified screen printed carbon electrode for sensing of amino acids. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114426] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
114
|
Webb L, Sadri H, Schuh K, Egert S, Stehle P, Meyer I, Koch C, Dusel G, Sauerwein H. Branched-chain amino acids: Abundance of their transporters and metabolizing enzymes in adipose tissue, skeletal muscle, and liver of dairy cows at high or normal body condition. J Dairy Sci 2020; 103:2847-2863. [DOI: 10.3168/jds.2019-17147] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
|
115
|
Chakravarthy MV, Waddell T, Banerjee R, Guess N. Nutrition and Nonalcoholic Fatty Liver Disease: Current Perspectives. Gastroenterol Clin North Am 2020; 49:63-94. [PMID: 32033765 DOI: 10.1016/j.gtc.2019.09.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis are diseases in their own right as well as modifiable risk factors for cardiovascular disease and type 2 diabetes. With expanding knowledge on NAFLD pathogenesis, insights have been gleaned into molecular targets for pharmacologic and nonpharmacologic approaches. Lifestyle modifications constitute a cornerstone of NAFLD management. This article reviews roles of key dietary macronutrients and micronutrients in NAFLD pathogenesis and their effects on molecular targets shared with established or emerging pharmacotherapies. Based on current evidence, a recommendation for a dietary framework as part of the comprehensive management strategy for NAFLD is provided.
Collapse
Affiliation(s)
| | - Thomas Waddell
- Perspectum Diagnostics, 23-38 Hythe Bridge Street, Oxford OX1 2ET, UK
| | - Rajarshi Banerjee
- Perspectum Diagnostics, 23-38 Hythe Bridge Street, Oxford OX1 2ET, UK; Oxford University Hospitals NHS Foundation Trust, Headley Way, Headington, Oxford OX3 9DU, UK
| | - Nicola Guess
- King's College London, 150 Stamford Street, London SE1 9NH, UK; University of Westminster, 101 New Cavendish St, Fitzrovia, London W1W 6XH, United Kingdom
| |
Collapse
|
116
|
Endotoxin Producers Overgrowing in Human Gut Microbiota as the Causative Agents for Nonalcoholic Fatty Liver Disease. mBio 2020; 11:mBio.03263-19. [PMID: 32019793 PMCID: PMC7002352 DOI: 10.1128/mbio.03263-19] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent studies have reported a link between gut microbiota and nonalcoholic fatty liver disease (NAFLD), showing that germfree (GF) mice do not develop metabolic syndromes, including NAFLD. However, the specific bacterial species causing NAFLD, as well as their molecular cross talk with the host for driving liver disease, remain elusive. Here, we found that nonvirulent endotoxin-producing strains of pathogenic species overgrowing in obese human gut can act as causative agents for induction of NAFLD and related metabolic disorders. The cross talk between endotoxin from these specific producers and the host’s TLR4 receptor is the most upstream and essential molecular event for inducing all phenotypes in NAFLD and related metabolic disorders. These nonvirulent endotoxin-producing strains of gut pathogenic species overgrowing in human gut may collectively become a predictive biomarker or serve as a novel therapeutic target for NAFLD and related metabolic disorders. Gut microbiota-derived endotoxin has been linked to human nonalcoholic fatty liver disease (NAFLD), but the specific causative agents and their molecular mechanisms remain elusive. In this study, we investigated whether bacterial strains of endotoxin-producing pathogenic species overgrowing in obese human gut can work as causative agents for NAFLD. We further assessed the role of lipopolysaccharide (LPS)-Toll-like receptor 4 (TLR4) cross talk in this pathogenicity. Nonvirulent strains of Gram-negative pathobionts were isolated from obese human gut and monoassociated with C57BL/6J germfree (GF) mice fed a high-fat diet (HFD). Deletion of waaG in the bacterial endotoxin synthetic pathway and knockout of TLR4 in GF mice were used to further study the underlying mechanism for a causal relationship between these strains and the development of NAFLD. Three endotoxin-producing strains, Enterobacter cloacae B29, Escherichia coli PY102, and Klebsiella pneumoniae A7, overgrowing in the gut of morbidly obese volunteers with severe fatty liver, induced NAFLD when monoassociated with GF mice on HFD, while HFD alone did not induce the disease in GF mice. The commensal Bacteroides thetaiotaomicron (ATCC 29148), whose endotoxin activity was markedly lower than that of Enterobacteriaceae strains, did not induce NAFLD in GF mice. B29 lost its proinflammatory properties and NAFLD-inducing capacity upon deletion of the waaG gene. Moreover, E. cloacae B29 did not induce NAFLD in TLR4-deficient GF mice. These nonvirulent endotoxin-producing strains in pathobiont species overgrowing in human gut may work as causative agents, with LPS-TLR4 cross talk as the most upstream and essential molecular event for NAFLD.
Collapse
|
117
|
Beyoğlu D, Idle JR. Metabolomic and Lipidomic Biomarkers for Premalignant Liver Disease Diagnosis and Therapy. Metabolites 2020; 10:E50. [PMID: 32012846 PMCID: PMC7074571 DOI: 10.3390/metabo10020050] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 01/24/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, there has been a plethora of attempts to discover biomarkers that are more reliable than α-fetoprotein for the early prediction and prognosis of hepatocellular carcinoma (HCC). Efforts have involved such fields as genomics, transcriptomics, epigenetics, microRNA, exosomes, proteomics, glycoproteomics, and metabolomics. HCC arises against a background of inflammation, steatosis, and cirrhosis, due mainly to hepatic insults caused by alcohol abuse, hepatitis B and C virus infection, adiposity, and diabetes. Metabolomics offers an opportunity, without recourse to liver biopsy, to discover biomarkers for premalignant liver disease, thereby alerting the potential of impending HCC. We have reviewed metabolomic studies in alcoholic liver disease (ALD), cholestasis, fibrosis, cirrhosis, nonalcoholic fatty liver (NAFL), and nonalcoholic steatohepatitis (NASH). Specificity was our major criterion in proposing clinical evaluation of indole-3-lactic acid, phenyllactic acid, N-lauroylglycine, decatrienoate, N-acetyltaurine for ALD, urinary sulfated bile acids for cholestasis, cervonoyl ethanolamide for fibrosis, 16α-hydroxyestrone for cirrhosis, and the pattern of acyl carnitines for NAFL and NASH. These examples derive from a large body of published metabolomic observations in various liver diseases in adults, adolescents, and children, together with animal models. Many other options have been tabulated. Metabolomic biomarkers for premalignant liver disease may help reduce the incidence of HCC.
Collapse
Affiliation(s)
| | - Jeffrey R. Idle
- Arthur G. Zupko’s Division of Systems Pharmacology and Pharmacogenomics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, 75 Dekalb Avenue, Brooklyn, NY 11201, USA;
| |
Collapse
|
118
|
Raigani S, Karimian N, Huang V, Zhang AM, Beijert I, Geerts S, Nagpal S, Hafiz EOA, Fontan FM, Aburawi MM, Mahboub P, Markmann JF, Porte RJ, Uygun K, Yarmush M, Yeh H. Metabolic and lipidomic profiling of steatotic human livers during ex situ normothermic machine perfusion guides resuscitation strategies. PLoS One 2020; 15:e0228011. [PMID: 31978172 PMCID: PMC6980574 DOI: 10.1371/journal.pone.0228011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/06/2020] [Indexed: 12/15/2022] Open
Abstract
There continues to be a significant shortage of donor livers for transplantation. One impediment is the discard rate of fatty, or steatotic, livers because of their poor post-transplant function. Steatotic livers are prone to significant ischemia-reperfusion injury (IRI) and data regarding how best to improve the quality of steatotic livers is lacking. Herein, we use normothermic (37°C) machine perfusion in combination with metabolic and lipidomic profiling to elucidate deficiencies in metabolic pathways in steatotic livers, and to inform strategies for improving their function. During perfusion, energy cofactors increased in steatotic livers to a similar extent as non-steatotic livers, but there were significant deficits in anti-oxidant capacity, efficient energy utilization, and lipid metabolism. Steatotic livers appeared to oxidize fatty acids at a higher rate but favored ketone body production rather than energy regeneration via the tricyclic acid cycle. As a result, lactate clearance was slower and transaminase levels were higher in steatotic livers. Lipidomic profiling revealed ω-3 polyunsaturated fatty acids increased in non-steatotic livers to a greater extent than in steatotic livers. The novel use of metabolic and lipidomic profiling during ex situ normothermic machine perfusion has the potential to guide the resuscitation and rehabilitation of steatotic livers for transplantation.
Collapse
Affiliation(s)
- Siavash Raigani
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Negin Karimian
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Viola Huang
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Anna M. Zhang
- Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Irene Beijert
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Sharon Geerts
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Sonal Nagpal
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Ehab O. A. Hafiz
- Electron Microscopy Research Division, Theodor Bilharz Research Institute, Giza, Egypt
| | - Fermin M. Fontan
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Mohamed M. Aburawi
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Paria Mahboub
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - James F. Markmann
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert J. Porte
- Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Korkut Uygun
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Martin Yarmush
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Engineering in Medicine, Massachusetts General Hospital and Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Heidi Yeh
- Division of Transplantation, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
119
|
Schumann T, König J, Henke C, Willmes DM, Bornstein SR, Jordan J, Fromm MF, Birkenfeld AL. Solute Carrier Transporters as Potential Targets for the Treatment of Metabolic Disease. Pharmacol Rev 2020; 72:343-379. [PMID: 31882442 DOI: 10.1124/pr.118.015735] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The solute carrier (SLC) superfamily comprises more than 400 transport proteins mediating the influx and efflux of substances such as ions, nucleotides, and sugars across biological membranes. Over 80 SLC transporters have been linked to human diseases, including obesity and type 2 diabetes (T2D). This observation highlights the importance of SLCs for human (patho)physiology. Yet, only a small number of SLC proteins are validated drug targets. The most recent drug class approved for the treatment of T2D targets sodium-glucose cotransporter 2, product of the SLC5A2 gene. There is great interest in identifying other SLC transporters as potential targets for the treatment of metabolic diseases. Finding better treatments will prove essential in future years, given the enormous personal and socioeconomic burden posed by more than 500 million patients with T2D by 2040 worldwide. In this review, we summarize the evidence for SLC transporters as target structures in metabolic disease. To this end, we identified SLC13A5/sodium-coupled citrate transporter, and recent proof-of-concept studies confirm its therapeutic potential in T2D and nonalcoholic fatty liver disease. Further SLC transporters were linked in multiple genome-wide association studies to T2D or related metabolic disorders. In addition to presenting better-characterized potential therapeutic targets, we discuss the likely unnoticed link between other SLC transporters and metabolic disease. Recognition of their potential may promote research on these proteins for future medical management of human metabolic diseases such as obesity, fatty liver disease, and T2D. SIGNIFICANCE STATEMENT: Given the fact that the prevalence of human metabolic diseases such as obesity and type 2 diabetes has dramatically risen, pharmacological intervention will be a key future approach to managing their burden and reducing mortality. In this review, we present the evidence for solute carrier (SLC) genes associated with human metabolic diseases and discuss the potential of SLC transporters as therapeutic target structures.
Collapse
Affiliation(s)
- Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Jörg König
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Stefan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Jens Jordan
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Martin F Fromm
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, Dresden University School of Medicine (T.S., C.H., D.M.W., S.R.B.), and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine (T.S., C.H., D.M.W.), Technische Universität Dresden, Dresden, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg, Germany (T.S., C.H., D.M.W., A.L.B.); Clinical Pharmacology and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany (J.K., M.F.F.); Institute for Aerospace Medicine, German Aerospace Center and Chair for Aerospace Medicine, University of Cologne, Cologne, Germany (J.J.); Diabetes and Nutritional Sciences, King's College London, London, United Kingdom (S.R.B., A.L.B.); Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany (A.L.B.); and Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, Eberhard Karls University Tübingen, Tübingen, Germany (A.L.B.)
| |
Collapse
|
120
|
Meda C, Barone M, Mitro N, Lolli F, Pedretti S, Caruso D, Maggi A, Della Torre S. Hepatic ERα accounts for sex differences in the ability to cope with an excess of dietary lipids. Mol Metab 2019; 32:97-108. [PMID: 32029233 PMCID: PMC6957843 DOI: 10.1016/j.molmet.2019.12.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Objective Among obesity-associated metabolic diseases, non-alcoholic fatty liver disease (NAFLD) represents an increasing public health issue due to its emerging association with atherogenic dyslipidemia and cardiovascular diseases (CVDs). The lower prevalence of NAFLD in pre-menopausal women compared with men or post-menopausal women led us to hypothesize that the female-inherent ability to counteract this pathology might strongly rely on estrogen signaling. In female mammals, estrogen receptor alpha (ERα) is highly expressed in the liver, where it acts as a sensor of the nutritional status and adapts the metabolism to the reproductive needs. As in the male liver this receptor is little expressed, we here hypothesize that hepatic ERα might account for sex differences in the ability of males and females to cope with an excess of dietary lipids and counteract the accumulation of lipids in the liver. Methods Through liver metabolomics and transcriptomics we analyzed the relevance of hepatic ERα in the metabolic response of males and females to a diet highly enriched in fats (HFD) as a model of diet-induced obesity. Results The study shows that the hepatic ERα strongly contributes to the sex-specific response to an HFD and its action accounts for opposite consequences for hepatic health in males and females. Conclusion This study identified hepatic ERα as a novel target for the design of sex-specific therapies against fatty liver and its cardio-metabolic consequences. Hepatic ERα contributes to sex-specific response to a fat-enriched diet. Hepatic ERα action accounts for contrasting consequences in males and females. In males, hepatic ERα action does not prevent liver lipid accumulation. The lack of ERα is responsible for an altered plasma lipid profile in males. In females, liver ERα controls lipid catabolism and counteracts NAFLD development.
Collapse
Affiliation(s)
- Clara Meda
- Department of Health Sciences, University of Milan, Italy
| | - Mara Barone
- Department of Pharmaceutical Sciences, University of Milan, Italy; Center of Excellence on Neurodegenerative Diseases, University of Milan, Italy
| | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Federica Lolli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Adriana Maggi
- Department of Pharmaceutical Sciences, University of Milan, Italy; Center of Excellence on Neurodegenerative Diseases, University of Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Italy; Center of Excellence on Neurodegenerative Diseases, University of Milan, Italy.
| |
Collapse
|
121
|
Gao J, Huang T, Li J, Guo X, Xiao H, Gu J, Tang J, Cai W, Li D. Beneficial Effects of n-3 Polyunsaturated Fatty Acids on Offspring's Pancreas of Gestational Diabetes Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:13269-13281. [PMID: 31725275 DOI: 10.1021/acs.jafc.9b05739] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
We studied the long-term influence of gestational diabetes mellitus (GDM) on the pancreas of offspring and the effect of omega-3 polyunsaturated fatty acids (n-3 PUFAs) on offspring's pancreas. GDM offspring were divided into three groups: GDM offspring, n-3 PUFA-adequate-GDM offspring, and n-3 PUFA-deficient GDM offspring. All healthy and GDM offspring were fed up to 11 months old. The pancreas of GDM offspring exhibited fatty infiltration at 11 months old, whereas n-3 PUFA improved the pancreatic fatty infiltration. n-3 PUFA lowered the pancreatic oxidative stress and inflammation. Surprisingly, n-3 PUFA postponed pancreatic telomere shortening of GDM offspring at old age. Nontargeted metabolomics showed that many metabolites were altered in the pancreas of GDM offspring at old age, including l-valine, ceramide, acylcarnitines, tocotrienol, cholesteryl acetate, and biotin. n-3 PUFA modulated some altered metabolites and metabolic pathways. Therefore, GDM caused the long-term effects on offspring's pancreas, whereas n-3 PUFA played a beneficial role.
Collapse
Affiliation(s)
- Jinlong Gao
- Department of Food Science and Nutrition , Zhejiang University , 866 Yuhangtang Road , Hangzhou 310058 , China
| | - Tao Huang
- Department of Epidemiology and Biostatistics , Peking University , 5 Yiheyuan Road , Beijing 100871 , China
| | - Jiaomei Li
- Institute of Nutrition and Health , Qingdao University , 308 Ningxia Road , Qingdao 266071 , China
| | - Xiaofei Guo
- Institute of Nutrition and Health , Qingdao University , 308 Ningxia Road , Qingdao 266071 , China
| | - Hailong Xiao
- Department of Food Inspection , Hangzhou Institute for Food and Drug Control , 198 Yonghua Street , Hangzhou 310022 , China
| | - Jiaojiao Gu
- School of Nursing , Zhejiang Chinese Medical University , 548 Binwen Road , Hangzhou 310053 , China
| | - Jun Tang
- Department of Food Science and Nutrition , Zhejiang University , 866 Yuhangtang Road , Hangzhou 310058 , China
| | - Wenwen Cai
- Department of Food Science and Nutrition , Zhejiang University , 866 Yuhangtang Road , Hangzhou 310058 , China
| | - Duo Li
- Department of Food Science and Nutrition , Zhejiang University , 866 Yuhangtang Road , Hangzhou 310058 , China
- Institute of Nutrition and Health , Qingdao University , 308 Ningxia Road , Qingdao 266071 , China
| |
Collapse
|
122
|
Sasaki GY, Li J, Cichon MJ, Riedl KM, Kopec RE, Bruno RS. Green Tea Extract Treatment in Obese Mice with Nonalcoholic Steatohepatitis Restores the Hepatic Metabolome in Association with Limiting Endotoxemia-TLR4-NFκB-Mediated Inflammation. Mol Nutr Food Res 2019; 63:e1900811. [PMID: 31574193 PMCID: PMC7293799 DOI: 10.1002/mnfr.201900811] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/18/2019] [Indexed: 12/15/2022]
Abstract
SCOPE Catechin-rich green tea extract (GTE) alleviates nonalcoholic steatohepatitis (NASH) by lowering endotoxin-TLR4 (Toll-like receptor-4)-NFκB (nuclear factor kappa-B) inflammation. This study aimed to define altered MS-metabolomic responses during high-fat (HF)-induced NASH that are restored by GTE utilizing livers from an earlier study in which GTE decreased endotoxin-TLR4-NFκB liver injury. METHODS AND RESULTS Mice are fed a low-fat (LF) or HF diet for 12 weeks and then randomized to LF or HF diets containing 0% or 2% GTE for an additional 8 weeks. Global MS-based metabolomics and targeted metabolite profiling of catechins/catechin metabolites are evaluated. GTE in HF mice restores hepatic metabolites implicated in dyslipidemia insulin resistance, and inflammation. These include 122 metabolites: amino acids, lipids, nucleotides, vitamins, bile acids, flavonoids, xenobiotics, and carbohydrates. Hepatic amino acids, B-vitamins, and bile acids are inversely correlated with biomarkers of insulin resistance, liver injury, steatosis, and inflammation. Further, phosphatidylcholine metabolites are positively correlated with biomarkers of liver injury and NFκB inflammation. Thirteen catechin metabolites are identified in livers of GTE-treated mice, mostly as phase II conjugates of parental catechins or microbial-derived valerolactones. CONCLUSION The defined anti-inflammatory/metabolic interactions advance an understanding of the mechanism by which GTE catechins protect against NFκB-mediated liver injury in NASH.
Collapse
Affiliation(s)
- Geoffrey Y Sasaki
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Jinhui Li
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Morgan J Cichon
- Personalized Food and Nutritional Metabolomics for Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Ken M Riedl
- Nutrient and Phytochemical Analytics Shared Resource, The Ohio State University, Columbus, OH, 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Rachel E Kopec
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
- Personalized Food and Nutritional Metabolomics for Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
123
|
Metabolic impact of partial hepatectomy in the non-alcoholic steatohepatitis animal model of methionine-choline deficient diet. J Pharm Biomed Anal 2019; 178:112958. [PMID: 31718984 DOI: 10.1016/j.jpba.2019.112958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 10/23/2019] [Accepted: 10/26/2019] [Indexed: 12/11/2022]
Abstract
In the liver, obesity is often manifested by the clinical disorder of the Non-Alcoholic Fatty Liver Disease (NAFLD). A proportion of NAFLD patients develop hepatic inflammation, known as Non-Alcoholic Steatohepatitis (NASH), which can end up in cirrhosis, or Hepatocellular Carcinoma (HCC). In this scenario, partial hepatectomy (PH) is an alternative to promote liver regeneration. However, as liver regeneration is impaired in NASH patients, more knowledge about its metabolic condition is needed to improve the regenerative response of the liver in this pathological condition. Although extensively employed, the panoply of molecular alterations involved in the regenerative response of the liver after partial hepatectomy PH is far from being fully characterized. Metabolic fingerprinting (metabolomics) is a powerful tool to help in the elucidation of complex metabolic networks, by means of a blind, naïve approach to study which metabolic nodes (metabolites) show the biggest variations between conditions. The objective of the present study was to gain deeper knowledge about the metabolic processes involved in the NASH animal model, and particularly in the effect of PH by using metabolomics. For achieving such information, twelve 8-week-old male C57BL/6 J mice, fed commercial chow (control diet) or methionine and choline-Deficient diet (MCD) for three weeks were subjected to PH and sacrificed 2 weeks later. Livers were removed and submitted to metabolic profiling analysis through RP-LC/MS (qTOF), GC/MS (qTOF) and CE/MS(TOF). More than 3000 different features were detected and repeated measurements one-way ANOVA analysis was performed to unveil significant features. MCD diet induced changes (p < 0.05) in 46% of the detected features, whereas PH provoked significant changes in 85% of them. Most of the changes were detected through LC/MS and were associated to lipid metabolism. However, changes of metabolites virtually related to other metabolic routes (amino acids, carbohydrates, nucleotides) were found altered and detected by CE/MS and GC/MS. The changes associated to PH show a similar trend regardless of the diet, but in the context of the diet deficient in methionine and choline we have found results that point to a different ratio glycolysis/tricarboxylic acid cycle. Moreover, in the NASH model, the regeneration of the liver structures occurs at the expense of an increased phosphatidylethanolamines/phosphatidylcholines ratio.
Collapse
|
124
|
Karusheva Y, Koessler T, Strassburger K, Markgraf D, Mastrototaro L, Jelenik T, Simon MC, Pesta D, Zaharia OP, Bódis K, Bärenz F, Schmoll D, Wolkersdorfer M, Tura A, Pacini G, Burkart V, Müssig K, Szendroedi J, Roden M. Short-term dietary reduction of branched-chain amino acids reduces meal-induced insulin secretion and modifies microbiome composition in type 2 diabetes: a randomized controlled crossover trial. Am J Clin Nutr 2019; 110:1098-1107. [PMID: 31667519 PMCID: PMC6821637 DOI: 10.1093/ajcn/nqz191] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/19/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Epidemiological studies have shown that increased circulating branched-chain amino acids (BCAAs) are associated with insulin resistance and type 2 diabetes (T2D). This may result from altered energy metabolism or dietary habits. OBJECTIVE We hypothesized that a lower intake of BCAAs improves tissue-specific insulin sensitivity. METHODS This randomized, placebo-controlled, double-blinded, crossover trial examined well-controlled T2D patients receiving isocaloric diets (protein: 1 g/kg body weight) for 4 wk. Protein requirements were covered by commercially available food supplemented ≤60% by an AA mixture either containing all AAs or lacking BCAAs. The dietary intervention ensured sufficient BCAA supply above the recommended minimum daily intake. The patients underwent the mixed meal tolerance test (MMT), hyperinsulinemic-euglycemic clamps (HECs), and skeletal muscle and white adipose tissue biopsies to assess insulin signaling. RESULTS After the BCAA- diet, BCAAs were reduced by 17% during fasting (P < 0.001), by 13% during HEC (P < 0.01), and by 62% during the MMT (P < 0.001). Under clamp conditions, whole-body and hepatic insulin sensitivity did not differ between diets. After the BCAA- diet, however, the oral glucose sensitivity index was 24% (P < 0.01) and circulating fibroblast-growth factor 21 was 21% higher (P < 0.05), whereas meal-derived insulin secretion was 28% lower (P < 0.05). Adipose tissue expression of the mechanistic target of rapamycin was 13% lower, whereas the mitochondrial respiratory control ratio was 1.7-fold higher (both P < 0.05). The fecal microbiome was enriched in Bacteroidetes but depleted of Firmicutes. CONCLUSIONS Short-term dietary reduction of BCAAs decreases postprandial insulin secretion and improves white adipose tissue metabolism and gut microbiome composition. Longer-term studies will be needed to evaluate the safety and metabolic efficacy in diabetes patients.This trial was registered at clinicaltrials.gov as NCT03261362.
Collapse
Affiliation(s)
- Yanislava Karusheva
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Theresa Koessler
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, München-Neuherberg, Germany,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Markgraf
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Lucia Mastrototaro
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Tomas Jelenik
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Marie-Christine Simon
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Dominik Pesta
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Oana-Patricia Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Felix Bärenz
- Sanofi-Aventis Deutschland GmbH, Frankfurt, Germany
| | | | | | - Andrea Tura
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
| | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padova, Italy
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany,Address correspondence to JS (e-mail: )
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany,German Center for Diabetes Research, München-Neuherberg, Germany,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
125
|
Softic S, Meyer JG, Wang GX, Gupta MK, Batista TM, Lauritzen HPMM, Fujisaka S, Serra D, Herrero L, Willoughby J, Fitzgerald K, Ilkayeva O, Newgard CB, Gibson BW, Schilling B, Cohen DE, Kahn CR. Dietary Sugars Alter Hepatic Fatty Acid Oxidation via Transcriptional and Post-translational Modifications of Mitochondrial Proteins. Cell Metab 2019; 30:735-753.e4. [PMID: 31577934 PMCID: PMC7816129 DOI: 10.1016/j.cmet.2019.09.003] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/06/2019] [Accepted: 09/05/2019] [Indexed: 01/25/2023]
Abstract
Dietary sugars, fructose and glucose, promote hepatic de novo lipogenesis and modify the effects of a high-fat diet (HFD) on the development of insulin resistance. Here, we show that fructose and glucose supplementation of an HFD exert divergent effects on hepatic mitochondrial function and fatty acid oxidation. This is mediated via three different nodes of regulation, including differential effects on malonyl-CoA levels, effects on mitochondrial size/protein abundance, and acetylation of mitochondrial proteins. HFD- and HFD plus fructose-fed mice have decreased CTP1a activity, the rate-limiting enzyme of fatty acid oxidation, whereas knockdown of fructose metabolism increases CPT1a and its acylcarnitine products. Furthermore, fructose-supplemented HFD leads to increased acetylation of ACADL and CPT1a, which is associated with decreased fat metabolism. In summary, dietary fructose, but not glucose, supplementation of HFD impairs mitochondrial size, function, and protein acetylation, resulting in decreased fatty acid oxidation and development of metabolic dysregulation.
Collapse
Affiliation(s)
- Samir Softic
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA 02115, USA; Division of Gastroenterology, Hepatology, Nutrition, Department of Pediatrics, University of Kentucky College of Medicine and Kentucky Children's Hospital, Lexington, KY 40506, USA.
| | - Jesse G Meyer
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Guo-Xiao Wang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Manoj K Gupta
- Islet Cell and Regenerative Medicine, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Thiago M Batista
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Hans P M M Lauritzen
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Shiho Fujisaka
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; First Department of Internal Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Dolors Serra
- School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Laura Herrero
- School of Pharmacy, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona 08028, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | | | | | - Olga Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC 27701, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Departments of Pharmacology & Cancer Biology and Medicine, Duke University Medical Center, Durham, NC 27701, USA
| | - Bradford W Gibson
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Birgit Schilling
- Chemistry & Mass Spectrometry, Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College New York, New York, NY 10021, USA
| | - C Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
126
|
Liu Z, Tu MJ, Zhang C, Jilek JL, Zhang QY, Yu AM. A reliable LC-MS/MS method for the quantification of natural amino acids in mouse plasma: Method validation and application to a study on amino acid dynamics during hepatocellular carcinoma progression. J Chromatogr B Analyt Technol Biomed Life Sci 2019; 1124:72-81. [PMID: 31177050 DOI: 10.1016/j.jchromb.2019.05.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/17/2019] [Accepted: 05/31/2019] [Indexed: 01/15/2023]
Abstract
A simple and fast LC-MS/MS method was developed and validated for simultaneous quantification of 20 proteinogenic l-amino acids (AAs) in a small volume (5 μL) of mouse plasma. Chromatographic separation was achieved on an Intrada Amino Acid column within 13 min via gradient elution with an aqueous solution containing 100 mM ammonium formate and an organic mobile phase containing acetonitrile, water and formic acid (v:v:v = 95:5:0.3), at the flow rate of 0.6 mL/min. Individual AAs and corresponding stable-isotope-labeled AAs internal standards were analyzed by multiple reaction monitoring (MRM) in positive ion mode under optimized conditions. Method validation consisted of linearity, sensitivity, accuracy and precision, recovery, matrix effect, and stability, and the results demonstrated this LC-MS/MS method as a specific, accurate, and reliable assay. This LC-MS/MS method was thus utilized to compare the dynamics of individual plasma AAs between healthy and orthotopic hepatocellular carcinoma (HCC) xenograft mice housed under identical conditions. Our results revealed that, 5 weeks after HCC tumor progression, plasma l-arginine concentrations were significantly decreased in HCC mice while l-alanine and l-threonine levels were sharply increased. These findings support the utilities of this LC-MS/MS method and the promise of specific AAs as possible biomarkers for HCC.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Department of Medical Function, Health Science Center, Yangtze University, Jingzhou, Hubei 434000, China; Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Mei-Juan Tu
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Chao Zhang
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Joseph L Jilek
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Qian-Yu Zhang
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA
| | - Ai-Ming Yu
- Department of Biochemistry & Molecular Medicine, School of Medicine, UC Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
127
|
Thoen RU, Barther NN, Schemitt E, Bona S, Fernandes S, Coral G, Marroni NP, Tovo C, Guedes RP, Porawski M. Zinc supplementation reduces diet-induced obesity and improves insulin sensitivity in rats. Appl Physiol Nutr Metab 2019; 44:580-586. [PMID: 30339765 DOI: 10.1139/apnm-2018-0519] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Rates of obesity have been growing at alarming rates, compromising the health of the world population. Thus, the search for interventions that address the metabolic repercussions of obesity are necessary. Here we evaluated the metabolic and antioxidant effects of zinc and branched-chain amino acids (BCAA) supplementation on obese rats. Male Wistar rats were fed either a high-fat/high-fructose diet (HFD) or a standard diet (SD) for 19 weeks. From the fifteenth week until the end of the experiment, HFD- and SD-fed rats received zinc (6 mg/kg) or BCAA (750 mg/kg) supplementation. Body weight, abdominal fat, lipid profile, blood glucose, insulin, leptin, and hepatic transaminases were evaluated. In the liver, superoxide dismutase and catalase activities and lipid peroxidation were also analyzed. HFD-fed animals showed increased weight gain, abdominal fat pad, plasma insulin, leptin, and triglycerides levels in comparison with SD-fed rats. Zinc supplementation reduced all these parameters, suggesting a beneficial role for the treatment of obesity. BCAA, on the other hand, did not show any beneficial effect. Liver antioxidant enzymes and hepatic transaminases plasma levels did not change among groups. Lipid peroxidation was higher in HFD-fed rats and was not reverted by zinc or BCAA supplementation. In conclusion, zinc supplementation may be a useful strategy for the treatment of the metabolic dysfunction associated with obesity.
Collapse
Affiliation(s)
- Rutiane Ullmann Thoen
- a Postgraduate Program in Medicine: Hepatology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS 90050-170, Brazil
| | - Nathaniele Nebel Barther
- a Postgraduate Program in Medicine: Hepatology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS 90050-170, Brazil
| | - Elizângela Schemitt
- b Postgraduate Program in Medical Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS 90035-007, Brazil
| | - Sílvia Bona
- b Postgraduate Program in Medical Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS 90035-007, Brazil
| | - Sabrina Fernandes
- a Postgraduate Program in Medicine: Hepatology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS 90050-170, Brazil
| | - Gabriela Coral
- a Postgraduate Program in Medicine: Hepatology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS 90050-170, Brazil
| | - Norma Possa Marroni
- b Postgraduate Program in Medical Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS 90035-007, Brazil
| | - Cristiane Tovo
- a Postgraduate Program in Medicine: Hepatology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS 90050-170, Brazil
| | - Renata Padilha Guedes
- c Postgraduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, RS 90050-170, Brazil
- d Postgraduate Program in Biosciences, UFCSPA, Porto Alegre, RS 90050-170, Brazil
| | - Marilene Porawski
- a Postgraduate Program in Medicine: Hepatology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, RS 90050-170, Brazil
- d Postgraduate Program in Biosciences, UFCSPA, Porto Alegre, RS 90050-170, Brazil
| |
Collapse
|
128
|
Untargeted metabolomic profiling of urine from healthy dogs and dogs with chronic hepatic disease. PLoS One 2019; 14:e0217797. [PMID: 31150490 PMCID: PMC6544284 DOI: 10.1371/journal.pone.0217797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/18/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic hepatic disease can present a diagnostic challenge with different etiologies being associated with similar clinical and laboratory findings. The histopathological assessment of a liver biopsy specimen is usually required in order to make a definitive diagnosis and the availability of non-invasive prognostic biomarkers is limited. The emerging science of metabolomics is used to detect changes in endogenous low molecular weight metabolites in biological samples and offers the possibility of identifying noninvasive markers of disease. The objective of this study was to investigate differences in the urine metabolome between healthy dogs, dogs with chronic hepatitis, dogs with hepatocellular carcinoma, and dogs with a congenital portosystemic shunt. Stored urine samples from 10 healthy dogs, 10 dogs with chronic hepatitis, 6 dogs with hepatocellular carcinoma, and 5 dogs with a congenital portosystemic shunt were analyzed. The urine metabolome was analyzed by gas chromatography–quadrupole time of flight mass spectrometry and 220 known metabolites were identified. Principal component analysis and heat dendrogram plots of the metabolomics data showed clustering between groups. Random forest analysis showed differences in the abundance of various metabolites including putrescine, gluconic acid, sorbitol, and valine. Based on univariate statistics, 37 metabolites were significantly different between groups. In, conclusion, the urine metabolome varies between healthy dogs, dogs with chronic hepatitis, dogs with hepatocellular carcinoma, and dogs with a congenital portosystemic shunt. Further targeted assessment of these metabolites is needed to assess their diagnostic utility.
Collapse
|
129
|
Biswas D, Duffley L, Pulinilkunnil T. Role of branched‐chain amino acid–catabolizing enzymes in intertissue signaling, metabolic remodeling, and energy homeostasis. FASEB J 2019; 33:8711-8731. [DOI: 10.1096/fj.201802842rr] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Dipsikha Biswas
- Department of Biochemistry and Molecular Biology Faculty of Medicine Dalhousie Medicine New Brunswick Dalhousie University Saint John New Brunswick Canada
| | - Luke Duffley
- Department of Biochemistry and Molecular Biology Faculty of Medicine Dalhousie Medicine New Brunswick Dalhousie University Saint John New Brunswick Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology Faculty of Medicine Dalhousie Medicine New Brunswick Dalhousie University Saint John New Brunswick Canada
| |
Collapse
|
130
|
Solon-Biet SM, Cogger VC, Pulpitel T, Wahl D, Clark X, Bagley E, Gregoriou GC, Senior AM, Wang QP, Brandon AE, Perks R, O’Sullivan J, Koay YC, Bell-Anderson K, Kebede M, Yau B, Atkinson C, Svineng G, Dodgson T, Wali JA, Piper MDW, Juricic P, Partridge L, Rose AJ, Raubenheimer D, Cooney GJ, Le Couteur DG, Simpson SJ. Branched chain amino acids impact health and lifespan indirectly via amino acid balance and appetite control. Nat Metab 2019; 1:532-545. [PMID: 31656947 PMCID: PMC6814438 DOI: 10.1038/s42255-019-0059-2] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/22/2019] [Indexed: 12/11/2022]
Abstract
Elevated branched chain amino acids (BCAAs) are associated with obesity and insulin resistance. How long-term dietary BCAAs impact late-life health and lifespan is unknown. Here, we show that when dietary BCAAs are varied against a fixed, isocaloric macronutrient background, long-term exposure to high BCAA diets leads to hyperphagia, obesity and reduced lifespan. These effects are not due to elevated BCAA per se or hepatic mTOR activation, but rather due to a shift in the relative quantity of dietary BCAAs and other AAs, notably tryptophan and threonine. Increasing the ratio of BCAAs to these AAs resulted in hyperphagia and is associated with central serotonin depletion. Preventing hyperphagia by calorie restriction or pair-feeding averts the health costs of a high BCAA diet. Our data highlight a role for amino acid quality in energy balance and show that health costs of chronic high BCAA intakes need not be due to intrinsic toxicity but, rather, a consequence of hyperphagia driven by AA imbalance.
Collapse
Affiliation(s)
- Samantha M Solon-Biet
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Victoria C Cogger
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
- Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord NSW, Australia
- ANZAC Research Institute, The University of Sydney NSW, Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Devin Wahl
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
- Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord NSW, Australia
| | - Ximonie Clark
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Elena Bagley
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Gabrielle C Gregoriou
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Medical Sciences, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Alistair M Senior
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Qiao-Ping Wang
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Guangzhou 510275, China
| | - Amanda E Brandon
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - Ruth Perks
- Charles Perkins Centre, The University of Sydney NSW, Australia
| | - John O’Sullivan
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Heart Research Institute, The University of Sydney, NSW, Australia
| | - Yen Chin Koay
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Heart Research Institute, The University of Sydney, NSW, Australia
| | - Kim Bell-Anderson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Melkam Kebede
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Belinda Yau
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Clare Atkinson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | | | - Timothy Dodgson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Jibran A Wali
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | | | - Paula Juricic
- Max Planck Institute for Biology of Aging, Cologne, Germany
| | | | - Adam J Rose
- Monash Biomedicine Discovery Institute, Monash University VIC, Australia
| | - David Raubenheimer
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| | - Gregory J Cooney
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
| | - David G Le Couteur
- Charles Perkins Centre, The University of Sydney NSW, Australia
- Sydney Medical School, Faculty of Health and Medicine, The University of Sydney NSW, Australia
- Ageing and Alzheimers Institute and Centre for Education and Research on Ageing, Concord Hospital, Concord NSW, Australia
- ANZAC Research Institute, The University of Sydney NSW, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, The University of Sydney NSW, Australia
- School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, NSW, Australia
| |
Collapse
|
131
|
Zhou W, Wang Y, Yang F, Dong Q, Wang H, Hu N. Rapid Determination of Amino Acids of Nitraria tangutorum Bobr. from the Qinghai-Tibet Plateau Using HPLC-FLD-MS/MS and a Highly Selective and Sensitive Pre-Column Derivatization Method. Molecules 2019; 24:E1665. [PMID: 31035340 PMCID: PMC6539371 DOI: 10.3390/molecules24091665] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 11/16/2022] Open
Abstract
Amino acids are indispensable components of living organisms. The high amino acid content in Nitraria tangutorum Bobr. fruit distinguishes it from other berry plants and is of great significance to its nutritional value. Herein, using 10-ethyl-acridine-3-sulfonyl chloride as a fluorescent pre-column labeling reagent, a method for the efficient and rapid determination of amino acid content in N. tangutorum by pre-column fluorescence derivatization and on-line mass spectrometry was established and further validated. The limits of detection (signal-to-noise ratio = 3) were between 0.13 and 1.13 nmol/L, with a linear coefficient greater than 0.997 and a relative standard deviation between 1.37% and 2.64%. In addition, the method required a short analysis time, separating 19 amino acids within 20 min. Subsequently, the method was used to analyze the amino acid content of Nitraria tangutorum Bobr. from tissues retrieved from seven regions of the Qinghai-Tibet Plateau. Nitraria tangutorum Bobr. was shown to contain a large amount of amino acids, with the total content and main amino acid varying between the different tissues. This research supports the nutritional evaluation, quality control, and development and utilization of Nitraria tangutorum Bobr.
Collapse
Affiliation(s)
- Wu Zhou
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China.
| | - Yuwei Wang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
| | - Fang Yang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining 810016, China.
| | - Qi Dong
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining 810008, China.
| | - Honglun Wang
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining 810008, China.
| | - Na Hu
- Key Laboratory of Tibetan Medicine Research, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining 810008, China.
- Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Xining 810008, China.
| |
Collapse
|
132
|
Non-Alcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: Role of Circulating Branched-Chain Amino Acids. Nutrients 2019; 11:nu11030705. [PMID: 30917546 PMCID: PMC6471562 DOI: 10.3390/nu11030705] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/14/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is likely to be associated with elevated plasma branched-chain amino acids (BCAAs) and may precede the development of type 2 diabetes (T2D). We hypothesized that BCAAs may be involved in the pathogenesis of T2D attributable to NAFLD and determined the extent to which plasma BCAAs influence T2D development in NAFLD. We evaluated cross-sectional associations of NAFLD with fasting plasma BCAAs (nuclear magnetic resonance spectroscopy), and prospectively determined the extent to which the influence of NAFLD on incident T2D is attributable to BCAA elevations. In the current study, 5791 Prevention of REnal and Vascular ENd-stage Disease (PREVEND) cohort participants without T2D at baseline were included. Elevated fatty liver index (FLI) ≥60, an algorithm based on triglycerides, gamma-glutamyltransferase, body mass index (BMI) and waist circumference, was used as proxy of NAFLD. Elevated FLI ≥ 60 was present in 1671 (28.9%) participants. Cross-sectionally, BCAAs were positively associated with FLI ≥ 60 (β = 0.208, p < 0.001). During a median follow-up of 7.3 years, 276 participants developed T2D, of which 194 (70.2%) had an FLI ≥ 60 (log-rank test, p < 0.001). Cox regression analyses revealed that both FLI ≥60 (hazard ratio (HR) 3.46, 95% CI 2.45⁻4.87, p < 0.001) and higher BCAA levels (HR 1.19, 95% CI 1.03⁻1.37, p = 0.01) were positively associated with incident T2D. Mediation analysis showed that the association of FLI with incident T2D was in part attributable to elevated BCAAs (proportion mediated 19.6%). In conclusion, both elevated FLI and elevated plasma BCAA levels are associated with risk of incident T2D. The association of NAFLD with T2D development seems partly mediated by elevated BCAAs.
Collapse
|
133
|
Mahmassani ZS, Reidy PT, McKenzie AI, Stubben C, Howard MT, Drummond MJ. Disuse-induced insulin resistance susceptibility coincides with a dysregulated skeletal muscle metabolic transcriptome. J Appl Physiol (1985) 2019; 126:1419-1429. [PMID: 30763167 DOI: 10.1152/japplphysiol.01093.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Short-term muscle disuse is characterized by skeletal muscle insulin resistance, although this response is divergent across subjects. The mechanisms regulating inactivity-induced insulin resistance between populations that are more or less susceptible to disuse-induced insulin resistance are not known. RNA sequencing was conducted on vastus lateralis muscle biopsies from subjects before and after bed rest (n = 26) to describe the transcriptome of inactivity-induced insulin resistance. Subjects were separated into Low (n = 14) or High (n = 12) Susceptibility Groups based on the magnitude of change in insulin sensitivity after 5 days of bed rest. Both groups became insulin-resistant after bed rest, and there were no differences between groups in nonmetabolic characteristics (body mass, body mass index, fat mass, and lean mass). The High Susceptibility Group had more genes altered >1.5-fold (426 high versus 391 low) and more than twofold (73 high versus 55 low). Twenty-four genes were altered more than twofold in the High Susceptibility Group that did not change in the Low Susceptibility Group. 95 gene changes correlated with the changes in insulin sensitivity; 6 of these genes changed more than twofold in the High Susceptibility Group. Participants in the High Susceptibility Group were uniquely characterized with muscle gene responses described by a decrease in pathways responsible for lipid uptake and oxidation, decreased capacity for triglyceride export (APOB), increased lipogenesis (i.e., PFKFB3, FASN), and increased amino acid export (SLC43A1). These transcriptomic data provide a comprehensive examination of pathways and genes that may be useful biomarkers, or novel targets to offset muscle disuse-induced insulin resistance. NEW & NOTEWORTHY Short-term muscle disuse results in skeletal muscle insulin resistance through mechanisms that are not fully understood. Following a 5-day bed rest intervention, subjects were divided into High and Low Susceptibility Groups to inactivity-induced insulin resistance. This was followed by a genome-wide transcriptional analysis on muscle biopsy samples to gain insight on divergent insulin sensitivity responses. Our primary finding was that the skeletal muscle of subjects who experienced the most inactivity-induced insulin resistance (high susceptibility) was characterized by a decreased preference for lipid oxidation, increased lipogenesis, and increased amino acid export.
Collapse
Affiliation(s)
- Ziad S Mahmassani
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| | - Paul T Reidy
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| | - Alec I McKenzie
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| | - Chris Stubben
- Bioinformatics Shared Resource at the Huntsman Cancer Institute , Salt Lake City, Utah
| | - Michael T Howard
- Department of Genetics, University of Utah , Salt Lake City, Utah
| | - Micah J Drummond
- Department of Physical Therapy and Athletic Training, University of Utah , Salt Lake City, Utah
| |
Collapse
|
134
|
Carulli L, Zanca G, Schepis F, Villa E. The OMICs Window into Nonalcoholic Fatty Liver Disease (NAFLD). Metabolites 2019; 9:25. [PMID: 30717274 PMCID: PMC6409793 DOI: 10.3390/metabo9020025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 12/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common cause of hepatic abnormalities worldwide. Nonalcoholic steatohepatitis (NASH) is part of the spectrum of NAFLD and leads to progressive liver disease, such as cirrhosis and hepatocellular carcinoma. In NASH patient, fibrosis represents the major predictor of liver-related mortality; therefore, it is important to have an early and accurate diagnosis of NASH. The current gold standard for the diagnosis of NASH is still liver biopsy. The development of biomarkers able to predict disease severity, prognosis, as well as response to therapy without the need for a biopsy is the focus of most up-to-date genomic, transcriptomic, proteomic, and metabolomic research. In the future, patients might be diagnosed and treated according to their molecular signatures. In this short review, we discuss how information from genomics, proteomics, and metabolomics contribute to the understanding of NAFLD pathogenesis.
Collapse
Affiliation(s)
- Lucia Carulli
- Division of Gastroenterology, Department of Medical Specialties, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Giulia Zanca
- Division of Gastroenterology, Department of Medical Specialties, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Filippo Schepis
- Division of Gastroenterology, Department of Medical Specialties, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Erica Villa
- Division of Gastroenterology, Department of Medical Specialties, University of Modena and Reggio Emilia, 41124 Modena, Italy.
| |
Collapse
|
135
|
Liu XJ, Zhang CE, Yu XH, Liu RX, Qin XM, Jia JD, Ma ZJ. Serum metabonomics characterization of liver fibrosis induced by bile duct-ligated in rats and the intervention effects of herb compound 861. J LIQ CHROMATOGR R T 2019. [DOI: 10.1080/10826076.2019.1574815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Xiao-Jie Liu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, P. R. China
| | - Cong-En Zhang
- Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Xiao-Hong Yu
- Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Rui-Xia Liu
- Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan, P. R. China
| | - Ji-Dong Jia
- Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis
| | - Zhi-Jie Ma
- Beijing Friendship Hospital, Capital Medical University, Beijing, P. R. China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis
| |
Collapse
|
136
|
A pilot study of the effect of phospholipid curcumin on serum metabolomic profile in patients with non-alcoholic fatty liver disease: a randomized, double-blind, placebo-controlled trial. Eur J Clin Nutr 2019; 73:1224-1235. [PMID: 30647436 DOI: 10.1038/s41430-018-0386-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND/OBJECTIVES Curcumin, a natural polyphenol compound in the spice turmeric, has been found to have potent anti-oxidative and anti-inflammatory activity. Curcumin may treat non-alcoholic fatty liver disease (NAFLD) through its beneficial effects on biomarkers of oxidative stress (OS) and inflammation, which are considered as two feature of this disease. However, the effects of curcumin on NAFLD have been remained poorly understood. This investigation evaluated the effects of administrating curcumin on metabolic status in NAFLD patients. SUBJECTS/METHODS Fifty-eight NAFLD patients participated in a randomized, double-blind, placebo-controlled parallel design of study. The subjects were allocated randomly into two groups, which either received 250 mg phospholipid curcumin or placebo, one capsule per day for a period of 8 weeks. Fasting blood samples were taken from each subject at the start and end of the study period. Subsequently, metabolomics analysis was performed for serum samples using NMR. RESULTS Compared with the placebo, supplementing phospholipid curcumin resulted in significant decreases in serum including 3- methyl-2-oxovaleric acid, 3-hydroxyisobutyrate, kynurenine, succinate, citrate, α-ketoglutarate, methylamine, trimethylamine, hippurate, indoxyl sulfate, chenodeoxycholic acid, taurocholic acid, and lithocholic acid. This profile of metabolic biomarkers could distinguish effectively NAFLD subjects who were treated with curcumin and placebo groups, achieving value of 0.99 for an area under receiver operating characteristic curve (AUC). CONCLUSIONS Characterizing the serum metabolic profile of the patients with NAFLD at the end of the intervention using NMR-based metabolomics method indicated that the targets of curcumin treatment included some amino acids, TCA cycle, bile acids, and gut microbiota.
Collapse
|
137
|
Gancheva S, Jelenik T, Álvarez-Hernández E, Roden M. Interorgan Metabolic Crosstalk in Human Insulin Resistance. Physiol Rev 2018; 98:1371-1415. [PMID: 29767564 DOI: 10.1152/physrev.00015.2017] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Excessive energy intake and reduced energy expenditure drive the development of insulin resistance and metabolic diseases such as obesity and type 2 diabetes mellitus. Metabolic signals derived from dietary intake or secreted from adipose tissue, gut, and liver contribute to energy homeostasis. Recent metabolomic studies identified novel metabolites and enlarged our knowledge on classic metabolites. This review summarizes the evidence of their roles as mediators of interorgan crosstalk and regulators of insulin sensitivity and energy metabolism. Circulating lipids such as free fatty acids, acetate, and palmitoleate from adipose tissue and short-chain fatty acids from the gut effectively act on liver and skeletal muscle. Intracellular lipids such as diacylglycerols and sphingolipids can serve as lipotoxins by directly inhibiting insulin action in muscle and liver. In contrast, fatty acid esters of hydroxy fatty acids have been recently shown to exert a series of beneficial effects. Also, ketoacids are gaining interest as potent modulators of insulin action and mitochondrial function. Finally, branched-chain amino acids not only predict metabolic diseases, but also inhibit insulin signaling. Here, we focus on the metabolic crosstalk in humans, which regulates insulin sensitivity and energy homeostasis in the main insulin-sensitive tissues, skeletal muscle, liver, and adipose tissue.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Tomas Jelenik
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Elisa Álvarez-Hernández
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| |
Collapse
|
138
|
A mix of dietary fermentable fibers improves lipids handling by the liver of overfed minipigs. J Nutr Biochem 2018; 65:72-82. [PMID: 30654277 DOI: 10.1016/j.jnutbio.2018.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/17/2018] [Accepted: 12/05/2018] [Indexed: 02/06/2023]
Abstract
Obesity induced by overfeeding ultimately can lead to nonalcoholic fatty liver disease, whereas dietary fiber consumption is known to have a beneficial effect. We aimed to determine if a supplementation of a mix of fibers (inulin, resistant starch and pectin) could limit or alleviate overfeeding-induced metabolic perturbations. Twenty female minipigs were fed with a control diet (C) or an enriched fat/sucrose diet supplemented (O + F) or not (O) with fibers. Between 0 and 56 days of overfeeding, insulin (+88%), HOMA (+102%), cholesterol (+45%) and lactate (+63%) were increased, without any beneficial effect of fibers supplementation. However, fibers supplementation limited body weight gain (vs. O, -15% at D56) and the accumulation of hepatic lipids droplets induced by overfeeding. This could be explained by a decreased lipids transport potential (-50% FABP1 mRNA, O + F vs. O) inducing a down-regulation of regulatory elements of lipids metabolism / lipogenesis (-36% SREBP1c mRNA, O + F vs. O) but not to an increased oxidation (O + F not different from O and C for proteins and mRNA measured). Glucose metabolism was also differentially regulated by fibers supplementation, with an increased net hepatic release of glucose in the fasted state (diet × time effect, P<.05 at D56) that can be explained partially by a possible increased glycogen synthesis in the fed state (+82% GYS2 protein, O + F vs. O, P=.09). The direct role of short chain fatty acids on gluconeogenesis stimulation is questioned, with probably a short-term impact (D14) but no effect on a long-term (D56) basis.
Collapse
|
139
|
Loomba R, Kayali Z, Noureddin M, Ruane P, Lawitz EJ, Bennett M, Wang L, Harting E, Tarrant JM, McColgan BJ, Chung C, Ray AS, Subramanian GM, Myers RP, Middleton MS, Lai M, Charlton M, Harrison SA. GS-0976 Reduces Hepatic Steatosis and Fibrosis Markers in Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology 2018; 155:1463-1473.e6. [PMID: 30059671 PMCID: PMC6318218 DOI: 10.1053/j.gastro.2018.07.027] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/18/2018] [Accepted: 07/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS De novo lipogenesis is increased in livers of patients with nonalcoholic steatohepatitis (NASH). Acetyl-coenzyme carboxylase catalyzes the rate-limiting step in this process. We evaluated the safety and efficacy of GS-0976, an inhibitor of acetyl-coenzyme A carboxylase in liver, in a phase 2 randomized placebo-controlled trial of patients with NASH. METHODS We analyzed data from 126 patients with hepatic steatosis of at least 8%, based on the magnetic resonance imaging-estimated proton density fat fraction (MRI-PDFF), and liver stiffness of at least 2.5 kPa, based on magnetic resonance elastography measurement or historical biopsy result consistent with NASH and F1-F3 fibrosis. Patients were randomly assigned (2:2:1) to groups given GS-0976 20 mg, GS-0976 5 mg, or placebo daily for 12 weeks, from August 8, 2016 through July 18, 2017. Measures of hepatic steatosis, stiffness, serum markers of fibrosis, and plasma metabolomics were evaluated. The primary aims were to confirm previous findings and evaluate the relation between dose and efficacy. RESULTS A relative decrease of at least 30% from baseline in MRI-PDFF (PDFF response) occurred in 48% of patients given GS-0976 20 mg (P = .004 vs placebo), 23% given GS-0976 5 mg (P = .43 vs placebo), and 15% given placebo. Median relative decreases in MRI-PDFF were greater in patients given GS-0976 20 mg (decrease of 29%) than those given placebo (decrease of 8%; P = .002). Changes in magnetic resonance elastography-measured stiffness did not differ among groups, but a dose-dependent decrease in the fibrosis marker tissue inhibitor of metalloproteinase 1 was observed in patients given GS-0976 20 mg. Plasma levels of acylcarnitine species also decreased in patients with a PDFF response given GS-0976 20 mg. GS-0976 was safe, but median relative increases of 11% and 13% in serum levels of triglycerides were observed in patients given GS-0976. CONCLUSIONS In a randomized placebo-controlled trial of patients with NASH, we found 12-week administration of GS-0976 20 mg decreased hepatic steatosis, selected markers of fibrosis, and liver biochemistry. ClinicalTrials.gov ID NCT02856555.
Collapse
Affiliation(s)
- Rohit Loomba
- University of California at San Diego, La Jolla, California.
| | - Zeid Kayali
- Inland Empire Liver Foundation, Rialto, California
| | | | - Peter Ruane
- Ruane Medical and Liver Health Institute, Los Angeles, California
| | - Eric J. Lawitz
- Texas Liver Institute, University of Texas Health San Antonio, San Antonio, Texas
| | - Michael Bennett
- Atlanta Gastroenterology Associates, Atlanta, Georgia; (6)Medical Research Associates Group, San Diego, California
| | - Lulu Wang
- Gilead Sciences, Inc, Foster City, California
| | | | | | | | | | | | | | | | | | - Michelle Lai
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | | |
Collapse
|
140
|
Pietzner M, Budde K, Homuth G, Kastenmüller G, Henning AK, Artati A, Krumsiek J, Völzke H, Adamski J, Lerch MM, Kühn JP, Nauck M, Friedrich N. Hepatic Steatosis Is Associated With Adverse Molecular Signatures in Subjects Without Diabetes. J Clin Endocrinol Metab 2018; 103:3856-3868. [PMID: 30060179 DOI: 10.1210/jc.2018-00999] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/24/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND AIMS Exaggerated hepatic triglyceride accumulation (i.e., hepatic steatosis) represents a strong risk factor for type 2 diabetes mellitus and cardiovascular disease. Despite the clear association of hepatic steatosis with impaired insulin signaling, the precise molecular mechanisms involved are still under debate. We combined data from several metabolomics techniques to gain a comprehensive picture of molecular alterations related to the presence of hepatic steatosis in a diabetes-free sample (N = 769) of the population-based Study of Health in Pomerania. METHODS Liver fat content (LFC) was assessed using MRI. Metabolome measurements of plasma and urine samples were done by mass spectrometry and nuclear magnetic resonance spectroscopy. Linear regression analyses were used to detect significant associations with either LFC or markers of hepatic damage. Possible mediations through insulin resistance, hypertriglyceridemia, and inflammation were tested. A predictive molecular signature of hepatic steatosis was established using regularized logistic regression. RESULTS The LFC-associated atherogenic lipid profile, tightly connected to shifts in the phospholipid content, and a prediabetic amino acid cluster were mediated by insulin resistance. Molecular surrogates of oxidative stress and multiple associations with urine metabolites (e.g., indicating altered cortisol metabolism or phase II detoxification products) were unaffected in mediation analyses. Incorporation of urine metabolites slightly improved classification of hepatic steatosis. CONCLUSIONS Comprehensive metabolic profiling allowed us to reveal molecular patterns accompanying hepatic steatosis independent of the known hallmarks. Novel biomarkers from urine (e.g., cortisol glucuronide) are worthwhile for follow-up in patients suffering from more severe liver impairment compared with our merely healthy population-based sample.
Collapse
Affiliation(s)
- Maik Pietzner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Kathrin Budde
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine and University Greifswald, Greifswald, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ann-Kristin Henning
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anna Artati
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jan Krumsiek
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Henry Völzke
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Diabetes Research (DZD), Site Greifswald, Greifswald, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Site Greifswald, Greifswald, Germany
- Lehrstuhl für Experimentelle Genetik, Technische Universität München, Freising-Weihenstephan, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Jens P Kühn
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
- Institute of Diagnostic Radiology, University Medicine, Carl Gustav Carus University, Dresden, Germany
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| |
Collapse
|
141
|
Wegermann K, Henao R, Diehl AM, Murphy SK, Abdelmalek MF, Moylan CA. Branched chain amino acid transaminase 1 (BCAT1) is overexpressed and hypomethylated in patients with non-alcoholic fatty liver disease who experience adverse clinical events: A pilot study. PLoS One 2018; 13:e0204308. [PMID: 30265706 PMCID: PMC6161885 DOI: 10.1371/journal.pone.0204308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 09/05/2018] [Indexed: 12/22/2022] Open
Abstract
Background and objectives Although the burden of non-alcoholic fatty liver disease (NAFLD) continues to increase worldwide, genetic factors predicting progression to cirrhosis and decompensation in NAFLD remain poorly understood. We sought to determine whether gene expression profiling was associated with clinical decompensation and death in patients with NAFLD, and to assess whether altered DNA methylation contributes to these changes in gene expression. Methods We performed a retrospective analysis of 86 patients in the Duke NAFLD Clinical Database and Biorepository with biopsy-proven NAFLD whose liver tissue was previously evaluated for gene expression and DNA methylation using array based technologies. We assessed the prospective development of liver and cardiovascular disease related outcomes, including hepatic decompensation as identified by the development of ascites, hepatic encephalopathy, hepatocellular carcinoma, or variceal bleeding as well as stroke and myocardial infarction via medical chart review. Results Of the 86 patients, 47 had F0-F1 fibrosis and 39 had F3-F4 fibrosis at index liver biopsy. Gene expression probe sets (n = 54,675) were analyzed; 42 genes showed significant differential expression (p<0.05) and a two-fold change in expression between patients with and without any outcome. Two expression probes of the branched chain amino-acid transaminase 1 (BCAT1) gene were upregulated (p = 0.02; fold change 2.1 and 2.2 respectively) in patients with a clinical outcome. Methylation of three of the 34 BCAT1 CpG methylation probes were significantly inversely correlated with BCAT1 expression specific to the probes predictive of clinical deterioration. Conclusion We found differential gene expression, correlated to changes in DNA methylation, at multiple BCAT1 loci in patients with cardiovascular outcomes and/or hepatic decompensation. BCAT1 catalyzes the transformation of alpha-ketoglutarate to glutamate and has been linked to the presence and severity of NAFLD, possibly through derangements in the balance between glutamate and alpha-ketoglutarate. Given the potential for BCAT1 to identify patients at risk for poor outcomes, and the potential therapeutic implications, these results should be validated in larger prospective studies.
Collapse
Affiliation(s)
- Kara Wegermann
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, United States of America
| | - Ricardo Henao
- Biostatistics and Bioinformatics, Duke University, Durham, NC, United States of America
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, United States of America
| | - Susan K. Murphy
- Department of Obstetrics and Gynecology, Duke University Health System, Durham, NC, United States of America
| | - Manal F. Abdelmalek
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, United States of America
| | - Cynthia A. Moylan
- Division of Gastroenterology, Department of Medicine, Duke University Health System, Durham, NC, United States of America
- Department of Medicine, Durham Veterans Affairs Health System, Durham, NC, United States of America
- * E-mail:
| |
Collapse
|
142
|
Baker PR, Friedman JE. Mitochondrial role in the neonatal predisposition to developing nonalcoholic fatty liver disease. J Clin Invest 2018; 128:3692-3703. [PMID: 30168806 DOI: 10.1172/jci120846] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global epidemic in obese children and adults, and the onset might have fetal origins. A growing body of evidence supports the role of developmental programming, whereby the maternal environment affects fetal and infant development, altering the risk profile for disease later in life. Human and nonhuman primate studies of maternal obesity demonstrate that risk factors for pediatric obesity and NAFLD begin in utero. The pathologic mechanisms for NAFLD are multifactorial but have centered on altered mitochondrial function/dysfunction that might precede insulin resistance. Compared with the adult liver, the fetal liver has fewer mitochondria, low activity of the fatty acid metabolic enzyme carnitine palmitoyl-CoA transferase-1, and little or no gluconeogenesis. Exposure to excess maternal fuels during fetal life uniquely alters hepatic fatty acid oxidation, tricarboxylic acid cycle activity, de novo lipogenesis, and mitochondrial health. These events promote increased oxidative stress and excess triglyceride storage, and, together with altered immune function and epigenetic changes, they prime the fetal liver for NAFLD and might drive the risk for nonalcoholic steatohepatitis in the next generation.
Collapse
Affiliation(s)
- Peter R Baker
- Section of Clinical Genetics and Metabolism, Department of Pediatrics
| | - Jacob E Friedman
- Section of Neonatology, Department of Pediatrics.,Department of Biochemistry and Molecular Genetics, and.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
143
|
Sano A, Kakazu E, Morosawa T, Inoue J, Kogure T, Ninomiya M, Iwata T, Umetsu T, Nakamura T, Takai S, Shimosegawa T. The profiling of plasma free amino acids and the relationship between serum albumin and plasma-branched chain amino acids in chronic liver disease: a single-center retrospective study. J Gastroenterol 2018; 53:978-988. [PMID: 29380062 DOI: 10.1007/s00535-018-1435-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND It is poorly understood how an imbalance of plasma-free amino acids (PFAAs) occurs and how the imbalance shows an association with the serum albumin (sAlb) level during the progression of chronic liver disease (CLDs). The aim of this study is to elucidate the profiles of PFAAs and the relationship between sAlb and PFAAs in recent patients with CLDs during the progression. METHODS We retrospectively evaluated the 1569 data of PFAAs data obtained from 908 patients with various CLDs (CHC, CHB. alcoholic, NAFLD/NASH, PBC, AIH, PSC, and cryptogenic). In total, 1140 data of PFAAs could be analyzed in patients with CLDs dependent of their Child-Pugh (CP) score. RESULTS Various imbalances in PFAAs were observed in each CLDs during the progression. Univariate and multivariate analysis revealed that among 24 PFAAs, the level of plasma-branched chain amino acids (pBCAAs) was significantly associated with the CP score, especially the sAlb score, in patients with chronic hepatitis C virus (CHC), NAFLD/NASH and PBC. The correlation coefficient values between sAlb and pBCAAs-to-Tyrosine ratio (BTR) in these patients were 0.53, 0.53 and 0.79, respectively. Interestingly, although the pBCAAs in NAFLD/NASH patients varied even when the sAlb was within the normal range, the pBCAAs tended to be low when the sAlb was below the normal range. CONCLUSIONS Although a decrease in the level of pBCAAs was observed during the progression regardless of the CLD etiology, the level of total pBCAAs was independently associated with the sAlb level in the PFAAs of CHC, PBC and NAFLD/NASH. The correlation between sAlb and BTR showed the highest value in PBC patients among the patients with CLDs. A decrease in pBCAAs often occurred in NASH even when the sAlb level was kept in the normal range.
Collapse
Affiliation(s)
- Akitoshi Sano
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Eiji Kakazu
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan.
| | - Tatsuki Morosawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Jun Inoue
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Takayuki Kogure
- Division of Gastroenterology, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aobaku, Sendai, Miyagi, 981-8558, Japan
| | - Masashi Ninomiya
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Tomoaki Iwata
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Teruyuki Umetsu
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Takuya Nakamura
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Satoshi Takai
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, 1-1 Seiryo, Aobaku, Sendai, 980-8574, Japan
| |
Collapse
|
144
|
Wang GE, Li YF, Zhai YJ, Gong L, Tian JY, Hong M, Yao N, Wu YP, Kurihara H, He RR. Theacrine protects against nonalcoholic fatty liver disease by regulating acylcarnitine metabolism. Metabolism 2018; 85:227-239. [PMID: 29727630 DOI: 10.1016/j.metabol.2018.04.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/27/2018] [Accepted: 04/29/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Acylcarnitine metabolism disorder contributes significantly to the pathogenesis of nonalcoholic fatty liver disease (NAFLD). There are, however, few ideal medications for NAFLD, which work by targeting acylcarnitine metabolism. The aim of this study was to investigate the protective effects of theacrine, a rare purine alkaloid isolated from Camellia assamica var. kucha, against acylcarnitine metabolism disorder in NAFLD. METHODS The pharmacological activities of theacrine were studied using high-fat diet (HFD)-fed ApoE-/- and C57BL/6J mice models. Oleate-treated HepG2 and L-02 cells were used to investigate the molecular mechanism of theacrine on acylcarnitine metabolism. The target of theacrine was confirmed in vitro as the blockade of sirtuin 3 (SIRT3) and protein kinase A. RESULTS Theacrine inhibits hepatic steatosis and liver inflammation and improves energy expenditure in HFD-fed mice. Theacrine ameliorates acylcarnitine metabolism disorder in HFD-fed mice and oleate-treated hepatocytes by improving fatty acid oxidation. The underlying mechanism involves theacrine's activation of the mitochondrial deacetylase SIRT3 and consequently, the increased activity of long-chain acyl coenzyme A dehydrogenase (LCAD) through deacetylation. CONCLUSION Theacrine promotes acylcarnitine metabolism in NAFLD through the SIRT3/LCAD signaling pathway. The target of theacrine's activities on NAFLD is identified as SIRT3.
Collapse
Affiliation(s)
- Guo-En Wang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yu-Jia Zhai
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Lian Gong
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jing-Yu Tian
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, Guangdong, China
| | - Mo Hong
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Nan Yao
- Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou 510095, China
| | - Yan-Ping Wu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Hiroshi Kurihara
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
145
|
Peng KY, Watt MJ, Rensen S, Greve JW, Huynh K, Jayawardana KS, Meikle PJ, Meex RCR. Mitochondrial dysfunction-related lipid changes occur in nonalcoholic fatty liver disease progression. J Lipid Res 2018; 59:1977-1986. [PMID: 30042157 DOI: 10.1194/jlr.m085613] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/19/2018] [Indexed: 12/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) comprises fat-accumulating conditions within hepatocytes that can cause severe liver damage and metabolic comorbidities. Studies suggest that mitochondrial dysfunction contributes to its development and progression and that the hepatic lipidome changes extensively in obesity and in NAFLD. To gain insight into the relationship between lipid metabolism and disease progression through different stages of NAFLD, we performed lipidomic analysis of plasma and liver biopsy samples from obese patients with nonalcoholic fatty liver (NAFL) or nonalcoholic steatohepatitis (NASH) and from those without NAFLD. Congruent with earlier studies, hepatic lipid levels overall increased with NAFLD. Lipid species that differed with NAFLD severity were related to mitochondrial dysfunction; specifically, hepatic cardiolipin and ubiquinone accumulated in NAFL, and levels of acylcarnitine increased with NASH. We propose that increased levels of cardiolipin and ubiquinone may help to preserve mitochondrial function in early NAFLD, but that mitochondrial function eventually fails with progression to NASH, leading to increased acylcarnitine. We also found a negative association between hepatic odd-chain phosphatidylcholine and NAFLD, which may result from mitochondrial dysfunction-related impairment of branched-chain amino acid catabolism. Overall, these data suggest a close link between accumulation of specific hepatic lipid species, mitochondrial dysfunction, and the progression of NAFLD.
Collapse
Affiliation(s)
- Kang-Yu Peng
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Matthew J Watt
- Department of Physiology, Faculty of Medicine, Dentistry, and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Sander Rensen
- Departments of Surgery Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Willem Greve
- Department of Surgery, Zuyderland Medical Center Heerlen, Heerlen, The Netherlands
| | - Kevin Huynh
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | | | - Peter J Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia .,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ruth C R Meex
- Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
146
|
Li X, Wu T, Jiang Y, Zhang Z, Han X, Geng W, Ding H, Kang J, Wang Q, Shang H. Plasma metabolic changes in Chinese HIV-infected patients receiving lopinavir/ritonavir based treatment: Implications for HIV precision therapy. Cytokine 2018; 110:204-212. [PMID: 29778008 DOI: 10.1016/j.cyto.2018.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/30/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVES The goal of this study is to profile the metabolic changes in the plasma of HIV patients receiving lopinavir/ritonavir (LPV/r)-based highly active antiretroviral therapy (HAART) relative to their treatment-naïve phase, aimed to identify precision therapy for HIV for improving prognosis and predicting dyslipidemia caused by LPV/r. METHODS 38 longitudinal plasma samples were collected from 19 HIV-infected patients both before and after antiretroviral therapy, and 18 samples from healthy individuals were used as controls. Untargeted metabolomics profiling of these plasma samples was performed using liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS). RESULTS A total of 331 compounds of known identity were detected among these metabolites, a 67-metabolite signature mainly mapping to tryptophan, histidine, acyl carnitine, ketone bodies and fatty acid metabolism distinguished HIV patients from healthy controls. The levels of 19 out of the 67 altered metabolites including histidine, kynurenine, and 3-hydroxybutyrate (BHBA), recovered after LPV/r-based antiretroviral therapy, and histidine was positively correlated with the presence of CD4 + T lymphocytes. Furthermore, using receiver operating characteristic (ROC) analyses, we discovered that butyrylcarnitine in combination with myristic acid from plasma in treatment-naïve patients could predict dyslipidemia caused by LPV/r with 87% accuracy. CONCLUSIONS Metabolites alterations in treatment-naïve HIV patients may indicate an inflammatory, oxidative state and mitochondrial dysfunction that is permissive for disease progression. Histidine may provide a specific protective function for HIV patients. Besides, elevated fatty acids levels including butyrylcarnitine and myristic acid after infection may indicate patients at risk of suffering from dyslipidemia after LPV/r-based HAART.
Collapse
Affiliation(s)
- Xiaolin Li
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Tong Wu
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Yongjun Jiang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Zining Zhang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Xiaoxu Han
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Wenqing Geng
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Haibo Ding
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Jing Kang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Qi Wang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China
| | - Hong Shang
- Key Laboratory of AIDS Immunology of National Health and Family Planning Commission, Department of Laboratory Medicine, The First Affiliated Hospital, China Medical University, Shenyang 110001, PR China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, PR China.
| |
Collapse
|
147
|
Fisel P, Schaeffeler E, Schwab M. Clinical and Functional Relevance of the Monocarboxylate Transporter Family in Disease Pathophysiology and Drug Therapy. Clin Transl Sci 2018; 11:352-364. [PMID: 29660777 PMCID: PMC6039204 DOI: 10.1111/cts.12551] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/27/2018] [Indexed: 12/14/2022] Open
Abstract
The solute carrier (SLC) SLC16 gene family comprises 14 members and encodes for monocarboxylate transporters (MCTs), which mediate the absorption and distribution of monocarboxylic compounds across plasma membranes. As the knowledge about their physiological function, activity, and regulation increases, their involvement and contribution to cancer and other diseases become increasingly evident. Moreover, promising opportunities for therapeutic interventions by directly targeting their endogenous functions or by exploiting their ability to deliver drugs to specific organ sites emerge.
Collapse
Affiliation(s)
- Pascale Fisel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tübingen, Tübingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tübingen, Tübingen, Germany.,Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany.,Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
148
|
Anhê FF, Varin TV, Le Barz M, Pilon G, Dudonné S, Trottier J, St-Pierre P, Harris CS, Lucas M, Lemire M, Dewailly É, Barbier O, Desjardins Y, Roy D, Marette A. Arctic berry extracts target the gut-liver axis to alleviate metabolic endotoxaemia, insulin resistance and hepatic steatosis in diet-induced obese mice. Diabetologia 2018; 61:919-931. [PMID: 29270816 DOI: 10.1007/s00125-017-4520-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 11/07/2017] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS There is growing evidence that fruit polyphenols exert beneficial effects on the metabolic syndrome, but the underlying mechanisms remain poorly understood. In the present study, we aimed to analyse the effects of polyphenolic extracts from five types of Arctic berries in a model of diet-induced obesity. METHODS Male C57BL/6 J mice were fed a high-fat/high-sucrose (HFHS) diet and orally treated with extracts of bog blueberry (BBE), cloudberry (CLE), crowberry (CRE), alpine bearberry (ABE), lingonberry (LGE) or vehicle (HFHS) for 8 weeks. An additional group of standard-chow-fed, vehicle-treated mice was included as a reference control for diet-induced obesity. OGTTs and insulin tolerance tests were conducted, and both plasma insulin and C-peptide were assessed throughout the OGTT. Quantitative PCR, western blot analysis and ELISAs were used to assess enterohepatic immunometabolic features. Faecal DNA was extracted and 16S rRNA gene-based analysis was used to profile the gut microbiota. RESULTS Treatment with CLE, ABE and LGE, but not with BBE or CRE, prevented both fasting hyperinsulinaemia (mean ± SEM [pmol/l]: chow 67.2 ± 12.3, HFHS 153.9 ± 19.3, BBE 114.4 ± 14.3, CLE 82.5 ± 13.0, CRE 152.3 ± 24.4, ABE 90.6 ± 18.0, LGE 95.4 ± 10.5) and postprandial hyperinsulinaemia (mean ± SEM AUC [pmol/l × min]: chow 14.3 ± 1.4, HFHS 31.4 ± 3.1, BBE 27.2 ± 4.0, CLE 17.7 ± 2.2, CRE 32.6 ± 6.3, ABE 22.7 ± 18.0, LGE 23.9 ± 2.5). None of the berry extracts affected C-peptide levels or body weight gain. Levels of hepatic serine phosphorylated Akt were 1.6-, 1.5- and 1.2-fold higher with CLE, ABE and LGE treatment, respectively, and hepatic carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-1 tyrosine phosphorylation was 0.6-, 0.7- and 0.9-fold increased in these mice vs vehicle-treated, HFHS-fed mice. These changes were associated with reduced liver triacylglycerol deposition, lower circulating endotoxins, alleviated hepatic and intestinal inflammation, and major gut microbial alterations (e.g. bloom of Akkermansia muciniphila, Turicibacter and Oscillibacter) in CLE-, ABE- and LGE-treated mice. CONCLUSIONS/INTERPRETATION Our findings reveal novel mechanisms by which polyphenolic extracts from ABE, LGE and especially CLE target the gut-liver axis to protect diet-induced obese mice against metabolic endotoxaemia, insulin resistance and hepatic steatosis, which importantly improves hepatic insulin clearance. These results support the potential benefits of these Arctic berries and their integration into health programmes to help attenuate obesity-related chronic inflammation and metabolic disorders. DATA AVAILABILITY All raw sequences have been deposited in the public European Nucleotide Archive server under accession number PRJEB19783 ( https://www.ebi.ac.uk/ena/data/view/PRJEB19783 ).
Collapse
Affiliation(s)
- Fernando F Anhê
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Bureau Y4340, Québec City, QC, G1V 4G5, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - Thibault V Varin
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - Mélanie Le Barz
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Bureau Y4340, Québec City, QC, G1V 4G5, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - Geneviève Pilon
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Bureau Y4340, Québec City, QC, G1V 4G5, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - Stéphanie Dudonné
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - Jocelyn Trottier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Centre, Laval University, Québec City, QC, Canada
- Faculty of Pharmacy, Laval University, Québec City, QC, Canada
| | - Philippe St-Pierre
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Bureau Y4340, Québec City, QC, G1V 4G5, Canada
| | - Cory S Harris
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Michel Lucas
- Populations Health and Optimal Health Practices Axis of the CHU-Québec Research Centre, Department of Social and Preventive Medicine, Laval University, Québec City, QC, Canada
| | - Mélanie Lemire
- Populations Health and Optimal Health Practices Axis of the CHU-Québec Research Centre, Department of Social and Preventive Medicine, Laval University, Québec City, QC, Canada
| | - Éric Dewailly
- Populations Health and Optimal Health Practices Axis of the CHU-Québec Research Centre, Department of Social and Preventive Medicine, Laval University, Québec City, QC, Canada
| | - Olivier Barbier
- Laboratory of Molecular Pharmacology, CHU-Québec Research Centre, Laval University, Québec City, QC, Canada
- Faculty of Pharmacy, Laval University, Québec City, QC, Canada
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - Denis Roy
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Bureau Y4340, Québec City, QC, G1V 4G5, Canada.
- Institute of Nutrition and Functional Foods, Laval University, Québec City, QC, Canada.
| |
Collapse
|
149
|
Gitto S, Schepis F, Andreone P, Villa E. Study of the Serum Metabolomic Profile in Nonalcoholic Fatty Liver Disease: Research and Clinical Perspectives. Metabolites 2018; 8:17. [PMID: 29495258 PMCID: PMC5876006 DOI: 10.3390/metabo8010017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/19/2018] [Accepted: 02/22/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, metabolomics has attracted great scientific attention. The metabolomics methodology might permit a view into transitional phases between healthy liver and nonalcoholic steatohepatitis. Metabolomics can help to analyze the metabolic alterations that play a main role in the progression of nonalcoholic steatohepatitis. Lipid, glucose, amino acid, and bile acid metabolism should be widely studied to understand the complex pathogenesis of nonalcoholic steatohepatitis. The discovery of new biomarkers would be important for diagnosis and staging of liver disease as well as for the assessment of efficacy of new drugs. Here, we review the metabolomics data regarding nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. We analyzed the main studies regarding the application of metabolomics methodology in the complex context of nonalcoholic steatohepatitis, trying to create a bridge from the basic to the clinical aspects.
Collapse
Affiliation(s)
- Stefano Gitto
- Department of Medical and Surgical Sciences, University of Bologna and Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, 40138 Bologna, Italy.
- Research Centre for the Study of Hepatitis, University of Bologna, 40138 Bologna, Italy.
| | - Filippo Schepis
- Department of Gastroenterology, Azienda Ospedaliero-Universitaria and University of Modena and Reggio Emilia, 41124 Modena, Italy.
| | - Pietro Andreone
- Department of Medical and Surgical Sciences, University of Bologna and Azienda Ospedaliero-Universitaria di Bologna, Policlinico Sant'Orsola-Malpighi, 40138 Bologna, Italy.
- Research Centre for the Study of Hepatitis, University of Bologna, 40138 Bologna, Italy.
| | - Erica Villa
- Department of Gastroenterology, Azienda Ospedaliero-Universitaria and University of Modena and Reggio Emilia, 41124 Modena, Italy.
| |
Collapse
|
150
|
Romero-Ibarguengoitia ME, Vadillo-Ortega F, Caballero AE, Ibarra-González I, Herrera-Rosas A, Serratos-Canales MF, León-Hernández M, González-Chávez A, Mummidi S, Duggirala R, López-Alvarenga JC. Family history and obesity in youth, their effect on acylcarnitine/aminoacids metabolomics and non-alcoholic fatty liver disease (NAFLD). Structural equation modeling approach. PLoS One 2018; 13:e0193138. [PMID: 29466466 PMCID: PMC5821462 DOI: 10.1371/journal.pone.0193138] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 02/05/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Structural equation modeling (SEM) can help understanding complex functional relationships among obesity, non-alcoholic fatty liver disease (NAFLD), family history of obesity, targeted metabolomics and pro-inflammatory markers. We tested two hypotheses: 1) If obesity precedes an excess of free fatty acids that increase oxidative stress and mitochondrial dysfunction, there would be an increase of serum acylcarnitines, amino acids and cytokines in obese subjects. Acylcarnitines would be related to non-alcoholic fatty disease that will induce insulin resistance. 2) If a positive family history of obesity and type 2 diabetes are the major determinants of the metabolomic profile, there would be higher concentration of amino acids and acylcarnitines in patients with this background that will induce obesity and NAFLD which in turn will induce insulin resistance. METHODS/RESULTS 137 normoglycemic subjects, mean age (SD) of 30.61 (8.6) years divided in three groups: BMI<25 with absence of NAFLD (G1), n = 82; BMI>30 with absence of NAFLD (G2), n = 24; and BMI>30 with NAFLD (G3), n = 31. Family history of obesity (any) was present in 53%. Both models were adjusted in SEM. Family history of obesity predicted obesity but could not predict acylcarnitines and amino acid concentrations (effect size <0.2), but did predict obesity phenotype. CONCLUSION Family history of obesity is the major predictor of obesity, and the metabolic abnormalities on amino acids, acylcarnitines, inflammation, insulin resistance, and NAFLD.
Collapse
Affiliation(s)
| | - Felipe Vadillo-Ortega
- Vinculation Unit Faculty of Medicine UNAM, Instituto Nacional de Medicina Genomica (INMEGEN), Mexico City, Mexico
| | | | | | | | | | | | | | - Srinivas Mummidi
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX, United States of America
| | - Ravindranath Duggirala
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX, United States of America
| | - Juan Carlos López-Alvarenga
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Edinburg, TX, United States of America
- Research department, Universidad Mexico Americana del Norte, Reynosa, Tamaulipas, Mexico
| |
Collapse
|