1
|
Pillet M, Dabrowski M, Marengo M, Fullgrabe L, Leduc M, Fontaine Q, Le Floch S, Huet V, Churlaud C, Lejeune P, Thomas H. Preliminary inter-port study of the quality of environments using physiological responses of invertebrates exposed to chronic trace element and organic contamination in Corsica (Mediterranean Sea). ECOTOXICOLOGY (LONDON, ENGLAND) 2023; 32:243-260. [PMID: 36797445 DOI: 10.1007/s10646-023-02635-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Port areas are socio-ecosystems impacted by chronic mixture pollution. Some marine species benefit from living there and may be studied to define the ecological state of such environments. In this study, the risks of chronic chemical contamination and its consequences on three marine molluscs were evaluated in North Corsica (France) port areas. Mediterranean mussel Mytilus galloprovincialis, tubular sea cucumber Holothuria tubulosa and Mediterranean limpet Patella sp. were sampled in three port areas and a reference location. A set of biomarkers was analysed to evaluate oxidative stress, detoxification, energetic metabolism, neurotoxicity, immunity and bioaccumulation (metallic trace elements and organic pollutants). The objectives were to assess pollution-induced effects in organisms, to determine the best bioindicator species for the selected locations and to validate a "pool" sampling technique (when the analysis is done on a single pool of samples and not on individual samples). The results validate the sampling techniques as "pool" for management purposes. St-Florent was demonstrated as the most contaminated location. All the other locations present a low contamination, below the recommended threshold values (for metallic trace elements and organic pollutants). Finally, the limpet appears to be the best bioindicator for the selected locations. Mussel and sea cucumber are inappropriate due to their absence in this oligotrophic region and the lack of responses observed, respectively.
Collapse
Affiliation(s)
- Marion Pillet
- LIttoral ENvironnement et Sociétés (UMR7266), La Rochelle Université, 02 rue Olympe de Gouges, 17000, La Rochelle, France.
- STAtion de REcherches Sous-marines et Océanographiques, Punta Revellata, BP33, 20260, Calvi, France.
| | - Mathilde Dabrowski
- LIttoral ENvironnement et Sociétés (UMR7266), La Rochelle Université, 02 rue Olympe de Gouges, 17000, La Rochelle, France
| | - Michel Marengo
- STAtion de REcherches Sous-marines et Océanographiques, Punta Revellata, BP33, 20260, Calvi, France
| | - Lovina Fullgrabe
- STAtion de REcherches Sous-marines et Océanographiques, Punta Revellata, BP33, 20260, Calvi, France
| | - Michèle Leduc
- STAtion de REcherches Sous-marines et Océanographiques, Punta Revellata, BP33, 20260, Calvi, France
| | - Quentin Fontaine
- STAtion de REcherches Sous-marines et Océanographiques, Punta Revellata, BP33, 20260, Calvi, France
| | - Stéphane Le Floch
- Centre de Documentation, de Recherche et d'Expérimentations sur les Pollutions Accidentelles des Eaux, 715 rue Alain Colas, CS 41836, 29218, Brest Cedex 2, France
| | - Valérie Huet
- LIttoral ENvironnement et Sociétés (UMR7266), La Rochelle Université, 02 rue Olympe de Gouges, 17000, La Rochelle, France
| | - Carine Churlaud
- LIttoral ENvironnement et Sociétés (UMR7266), La Rochelle Université, 02 rue Olympe de Gouges, 17000, La Rochelle, France
| | - Pierre Lejeune
- STAtion de REcherches Sous-marines et Océanographiques, Punta Revellata, BP33, 20260, Calvi, France
| | - Hélène Thomas
- LIttoral ENvironnement et Sociétés (UMR7266), La Rochelle Université, 02 rue Olympe de Gouges, 17000, La Rochelle, France
| |
Collapse
|
2
|
Zhang K, Zhao W, Rodionov DA, Rubinstein GM, Nguyen DN, Tanwee TNN, Crosby J, Bing RG, Kelly RM, Adams MWW, Zhang Y. Genome-Scale Metabolic Model of Caldicellulosiruptor bescii Reveals Optimal Metabolic Engineering Strategies for Bio-based Chemical Production. mSystems 2021; 6:e0135120. [PMID: 34060912 PMCID: PMC8269263 DOI: 10.1128/msystems.01351-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/04/2021] [Indexed: 12/03/2022] Open
Abstract
Metabolic modeling was used to examine potential bottlenecks that could be encountered for metabolic engineering of the cellulolytic extreme thermophile Caldicellulosiruptor bescii to produce bio-based chemicals from plant biomass. The model utilizes subsystems-based genome annotation, targeted reconstruction of carbohydrate utilization pathways, and biochemical and physiological experimental validations. Specifically, carbohydrate transport and utilization pathways involving 160 genes and their corresponding functions were incorporated, representing the utilization of C5/C6 monosaccharides, disaccharides, and polysaccharides such as cellulose and xylan. To illustrate its utility, the model predicted that optimal production from biomass-based sugars of the model product, ethanol, was driven by ATP production, redox balancing, and proton translocation, mediated through the interplay of an ATP synthase, a membrane-bound hydrogenase, a bifurcating hydrogenase, and a bifurcating NAD- and NADP-dependent oxidoreductase. These mechanistic insights guided the design and optimization of new engineering strategies for product optimization, which were subsequently tested in the C. bescii model, showing a nearly 2-fold increase in ethanol yields. The C. bescii model provides a useful platform for investigating the potential redox controls that mediate the carbon and energy flows in metabolism and sets the stage for future design of engineering strategies aiming at optimizing the production of ethanol and other bio-based chemicals. IMPORTANCE The extremely thermophilic cellulolytic bacterium, Caldicellulosiruptor bescii, degrades plant biomass at high temperatures without any pretreatments and can serve as a strategic platform for industrial applications. The metabolic engineering of C. bescii, however, faces potential bottlenecks in bio-based chemical productions. By simulating the optimal ethanol production, a complex interplay between redox balancing and the carbon and energy flow was revealed using a C. bescii genome-scale metabolic model. New engineering strategies were designed based on an improved mechanistic understanding of the C. bescii metabolism, and the new designs were modeled under different genetic backgrounds to identify optimal strategies. The C. bescii model provided useful insights into the metabolic controls of this organism thereby opening up prospects for optimizing production of a wide range of bio-based chemicals.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Cell and Molecular Biology, College of the Environment and Life Sciences, University of Rhode Island, Kingston, Rhode Island, USA
| | - Weishu Zhao
- Department of Cell and Molecular Biology, College of the Environment and Life Sciences, University of Rhode Island, Kingston, Rhode Island, USA
| | - Dmitry A. Rodionov
- Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, California, USA
- A.A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, Russia
| | - Gabriel M. Rubinstein
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Diep N. Nguyen
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Tania N. N. Tanwee
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - James Crosby
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Ryan G. Bing
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Robert M. Kelly
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Michael W. W. Adams
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Ying Zhang
- Department of Cell and Molecular Biology, College of the Environment and Life Sciences, University of Rhode Island, Kingston, Rhode Island, USA
| |
Collapse
|
3
|
Oliva ME, Ferreira MDR, Vega Joubert MB, D'Alessandro ME. Salvia hispanica L. (chia) seed promotes body fat depletion and modulates adipocyte lipid handling in sucrose-rich diet-fed rats. Food Res Int 2021; 139:109842. [PMID: 33509466 DOI: 10.1016/j.foodres.2020.109842] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/13/2020] [Accepted: 10/18/2020] [Indexed: 11/16/2022]
Abstract
The aim of this study was to analyze the effects of Salvia hispanica L. (chia) seed upon metabolic pathways that play a key role in adipose tissue lipid handling which could be involved in visceral adiposity reduction developed in rats fed a sucrose-rich diet (SRD). Male Wistar rats were fed with a reference diet (RD) -6 months- or SRD-3 months. Then, the last group was randomly divided into two subgroups. One subgroup continued receiving the SRD up to 6 months and the other was fed with a SRD where whole chia seed was incorporated as the source of dietary fat for the next 3 months (SRD + CHIA). Results showed that chia seed in the SRD-fed rat reduced the abdominal and thoracic circumferences, carcass fat content, adipose tissue weights, and visceral adiposity index. This was accompanied by an improvement in insulin sensitivity and plasma lipid profile. In epididymal adipose tissue, the decreased fat cell triglyceride content was associated with a reduction in both, FAT/CD 36 plasma membrane levels and the fat synthesis enzyme activities. There were not changes in oxidative CPT enzyme activities. PKCβ and the precursor and mature forms of SREBP-1 protein levels were decreased, while pAMPK was increased. Our findings suggest that chia seed supplementation can modulate essential pathways of lipid metabolism in adipose tissue, contributing to reduced visceral fat accumulation in SRD-fed rats.
Collapse
Affiliation(s)
- María Eugenia Oliva
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - María Del Rosario Ferreira
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Michelle Berenice Vega Joubert
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - María Eugenia D'Alessandro
- Laboratorio de Estudio de Enfermedades Metabólicas relacionadas con la Nutrición, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Ciudad Universitaria, Santa Fe, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
4
|
de Toro-Martín J, Fernández-Marcelo T, González-Rodríguez Á, Escrivá F, Valverde ÁM, Álvarez C, Fernández-Millán E. Defective liver glycogen autophagy related to hyperinsulinemia in intrauterine growth-restricted newborn wistar rats. Sci Rep 2020; 10:17651. [PMID: 33077861 PMCID: PMC7573689 DOI: 10.1038/s41598-020-74702-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
Maternal malnutrition plays a critical role in the developmental programming of later metabolic diseases susceptibility in the offspring, such as obesity and type 2 diabetes. Because the liver is the major organ that produces and supplies blood glucose, we aimed at defining the potential role of liver glycogen autophagy in the programming of glucose metabolism disturbances. To this end, newborns were obtained from pregnant Wistar rats fed ad libitum with a standard diet or 65% food-restricted during the last week of gestation. We found that newborns from undernourished mothers showed markedly high basal insulin levels whereas those of glucagon were decreased. This unbalance led to activation of the mTORC1 pathway and inhibition of hepatic autophagy compromising the adequate handling of glycogen in the very early hours of extrauterine life. Restoration of autophagy with rapamycin but not with glucagon, indicated no defect in autophagy machinery per se, but in signals triggered by glucagon. Taken together, these results support the notion that hyperinsulinemia is an important mechanism by which mobilization of liver glycogen by autophagy is defective in food-restricted animals. This early alteration in the hormonal control of liver glycogen autophagy may influence the risk of developing metabolic diseases later in life.
Collapse
Affiliation(s)
- Juan de Toro-Martín
- Centre Nutrition, Santé et Société (NUTRISS)-Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Québec City, QC, Canada
| | - Tamara Fernández-Marcelo
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029, Madrid, Spain
| | - Águeda González-Rodríguez
- Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Madrid, Spain
| | - Fernando Escrivá
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029, Madrid, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, UCM, Ciudad Universitaria s/n, 28040, Madrid, Spain
| | - Ángela M Valverde
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029, Madrid, Spain.,Instituto de Investigaciones Biomédicas Alberto Sols (IIBm) (CSIC/UAM), C/ Arturo Duperier 4, 28029, Madrid, Spain
| | - Carmen Álvarez
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029, Madrid, Spain. .,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, UCM, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| | - Elisa Fernández-Millán
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), ISCIII, 28029, Madrid, Spain. .,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, UCM, Ciudad Universitaria s/n, 28040, Madrid, Spain.
| |
Collapse
|
5
|
Kosenko E, Tikhonova L, Alilova G, Montoliu C. A Look into Liver Mitochondrial Dysfunction as a Hallmark in Progression of Brain Energy Crisis and Development of Neurologic Symptoms in Hepatic Encephalopathy. J Clin Med 2020; 9:E2259. [PMID: 32708652 PMCID: PMC7408643 DOI: 10.3390/jcm9072259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/18/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The relationship between liver disease and neuropathology in hepatic encephalopathy is well known, but the genesis of encephalopathy in liver failure is yet to be elucidated. Conceptually, the main cause of hepatic encephalopathy is the accumulation of brain ammonia due to impaired liver detoxification function or occurrence of portosystemic shunt. Yet, as well as taking up toxic ammonia, the liver also produces vital metabolites that ensure normal cerebral function. Given this, for insight into how perturbations in the metabolic capacity of the liver may be related to brain pathology, it is crucial to understand the extent of ammonia-related changes in the hepatic metabolism that provides respiratory fuel for the brain, a deficiency of which can give rise to encephalopathy. METHODS Hepatic encephalopathy was induced in starved rats by injection of ammonium acetate. Ammonia-induced toxicity was evaluated by plasma and freeze-clamped liver and brain energy metabolites, and mitochondrial, cytoplasmic, and microsomal gluconeogenic enzymes, including mitochondrial ketogenic enzymes. Parameters of oxidative phosphorylation were recorded polarographically with a Clark-type electrode, while other measures were determined with standard fluorometric enzymatic methods. RESULTS Progressive impairment of liver mitochondrial respiration in the initial stage of ammonia-induced hepatotoxicity and the subsequent energy crisis due to decreased ATP synthesis lead to cessation of gluconeogenesis and ketogenesis. Reduction in glucose and ketone body supply to the brain is a terminal event in liver toxicity, preceding the development of coma. CONCLUSIONS Our study provides a framework to further explore the relationship between hepatic dysfunction and progression of brain energy crisis in hepatic encephalopathy.
Collapse
Affiliation(s)
- Elena Kosenko
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia; (L.T.); (G.A.)
| | - Lyudmila Tikhonova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia; (L.T.); (G.A.)
| | - Gubidat Alilova
- Institute of Theoretical and Experimental Biophysics of Russian Academy of Sciences, 142290 Pushchino, Russia; (L.T.); (G.A.)
| | - Carmina Montoliu
- Hospital Clinico Research Foundation, INCLIVA Health Research Institute, 46010 Valencia, Spain;
- Pathology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
6
|
Yang X, Zhao Y, Sun Q, Yang Y, Gao Y, Ge W, Liu J, Xu X, Weng D, Wang S, Zhang J. Adenine nucleotide-mediated regulation of hepatic PTP1B activity in mouse models of type 2 diabetes. Diabetologia 2019; 62:2106-2117. [PMID: 31410531 DOI: 10.1007/s00125-019-04971-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/24/2019] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Plasma 5'-AMP (pAMP) is elevated in mouse models of type 2 diabetes. However, the metabolic regulatory role of adenine nucleotides in type 2 diabetes remains unclear. METHODS Adenine nucleotides and their metabolites in plasma and liver were examined by HPLC. 1H NMR-based metabolomics analysis was performed to explore the changes of metabolites in mouse models of type 2 diabetes. Na+/K+ ATPase and Na+/H+ exchanger activity were measured in response to adenine nucleotide metabolites. Human recombinant protein tyrosine phosphatase 1B (PTP1B) was used for enzyme kinetic assays. Protein binding assays were performed with microscale thermophoresis. The intracellular pH of hepatocyte AML12 cell lines was measured using the BCECF-AM method. We also analysed pAMP levels in participants with type 2 diabetes. RESULTS Elevation of pAMP was a universal phenomenon in all mouse models of type 2 diabetes including db/db vs lean mice (13.9 ± 2.3 μmol/l vs 3.7 ± 0.9 μmol/l; p < 0.01), ob/ob vs lean mice (9.1 ± 2.0 μmol/l vs 3.9 ± 1.2 μmol/l; p < 0.01) and high-fat diet/streptozotocin-induced vs wild-type mice (6.6 ± 1.5 μmol/l vs 4.1 ± 0.9 μmol/l; p < 0.05); this elevation was required for the occurrence of hyperglycaemia in obese mice. 1H NMR-based metabolomics study following HPLC analysis revealed that the metabolite profile in wild-type mice treated with 5'-AMP was similar to that in db/db diabetic mice, especially the accumulation of a large quantity of ATP and its metabolites. The glucose-lowering drug metformin reduced the severity of hyperglycaemia both in 5'-AMP-induced wild-type mice and db/db mice. Metformin decreased the accumulation of liver ATP but not its metabolites in these hyperglycaemic mice. ATP and metformin reciprocally change cellular pH homeostasis in liver, causing opposite shifts in liver activity of PTP1B, a key negative regulator of insulin signalling. Furthermore, pAMP levels were also elevated in individuals with type 2 diabetes (45.2 ± 22.7 nmol/l vs 3.1 ± 1.9 nmol/l; p < 0.01). CONCLUSIONS/INTERPRETATION These results reveal an emerging role for adenine nucleotide in the regulation of hyperglycaemia and provide a potential therapeutic target in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Xiao Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Yang Zhao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Qi Sun
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Yunxia Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Yan Gao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Wenhao Ge
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Junhao Liu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Dan Weng
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Shiming Wang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiaolingwei, Nanjing, 210094, China.
| |
Collapse
|
7
|
Hunter RW, Hughey CC, Lantier L, Sundelin EI, Peggie M, Zeqiraj E, Sicheri F, Jessen N, Wasserman DH, Sakamoto K. Metformin reduces liver glucose production by inhibition of fructose-1-6-bisphosphatase. Nat Med 2018; 24:1395-1406. [PMID: 30150719 PMCID: PMC6207338 DOI: 10.1038/s41591-018-0159-7] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 07/24/2018] [Indexed: 01/03/2023]
Abstract
Metformin is a first-line drug for the treatment of individuals with type 2 diabetes, yet its precise mechanism of action remains unclear. Metformin exerts its antihyperglycemic action primarily through lowering hepatic glucose production (HGP). This suppression is thought to be mediated through inhibition of mitochondrial respiratory complex I, and thus elevation of 5'-adenosine monophosphate (AMP) levels and the activation of AMP-activated protein kinase (AMPK), though this proposition has been challenged given results in mice lacking hepatic AMPK. Here we report that the AMP-inhibited enzyme fructose-1,6-bisphosphatase-1 (FBP1), a rate-controlling enzyme in gluconeogenesis, functions as a major contributor to the therapeutic action of metformin. We identified a point mutation in FBP1 that renders it insensitive to AMP while sparing regulation by fructose-2,6-bisphosphate (F-2,6-P2), and knock-in (KI) of this mutant in mice significantly reduces their response to metformin treatment. We observe this during a metformin tolerance test and in a metformin-euglycemic clamp that we have developed. The antihyperglycemic effect of metformin in high-fat diet-fed diabetic FBP1-KI mice was also significantly blunted compared to wild-type controls. Collectively, we show a new mechanism of action for metformin and provide further evidence that molecular targeting of FBP1 can have antihyperglycemic effects.
Collapse
Affiliation(s)
- Roger W Hunter
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland
- School of Physiology, Pharmacology & Neuroscience, University of Bristol, Bristol, UK
| | - Curtis C Hughey
- Department of Molecular Physiology and Biophysics and the Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics and the Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN, USA
| | - Elias I Sundelin
- Departments of Clinical Medicine and Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mark Peggie
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Elton Zeqiraj
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Frank Sicheri
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Departments of Biochemistry and Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Niels Jessen
- Departments of Clinical Medicine and Biomedicine, Aarhus University, Aarhus, Denmark
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics and the Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University, Nashville, TN, USA
| | - Kei Sakamoto
- Nestlé Institute of Health Sciences SA, Lausanne, Switzerland.
| |
Collapse
|
8
|
Dong S, Baranwal S, Garcia A, Serrano-Gomez SJ, Eastlack S, Iwakuma T, Mercante D, Mauvais-Jarvis F, Alahari SK. Nischarin inhibition alters energy metabolism by activating AMP-activated protein kinase. J Biol Chem 2017; 292:16833-16846. [PMID: 28842496 DOI: 10.1074/jbc.m117.784256] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 08/22/2017] [Indexed: 11/06/2022] Open
Abstract
Nischarin (Nisch) is a key protein functioning as a molecular scaffold and thereby hosting interactions with several protein partners. To explore the physiological importance of Nisch, here we generated Nisch loss-of-function mutant mice and analyzed their metabolic phenotype. Nisch-mutant embryos exhibited delayed development, characterized by small size and attenuated weight gain. We uncovered the reason for this phenotype by showing that Nisch binds to and inhibits the activity of AMP-activated protein kinase (AMPK), which regulates energy homeostasis by suppressing anabolic and activating catabolic processes. The Nisch mutations enhanced AMPK activation and inhibited mechanistic target of rapamycin signaling in mouse embryonic fibroblasts as well as in muscle and liver tissues of mutant mice. Nisch-mutant mice also exhibited increased rates of glucose oxidation with increased energy expenditure, despite reduced overall food intake. Moreover, the Nisch-mutant mice had reduced expression of liver markers of gluconeogenesis associated with increased glucose tolerance. As a result, these mice displayed decreased growth and body weight. Taken together, our results indicate that Nisch is an important AMPK inhibitor and a critical regulator of energy homeostasis, including lipid and glucose metabolism.
Collapse
Affiliation(s)
- Shengli Dong
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and
| | - Somesh Baranwal
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and.,the Center for Biochemistry and Microbial Sciences, Central University of Punjab, City Campus Mansa Rd., Bathinda-151001, India
| | - Anapatricia Garcia
- the Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia 30322
| | - Silvia J Serrano-Gomez
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and.,the Pontificia Universidad Javeriana, 11001000 Bogotá, Colombia
| | - Steven Eastlack
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and
| | - Tomoo Iwakuma
- the Department of Cancer Biology, Kansas University Medical Center, Kansas City, Kansas 66160, and
| | - Donald Mercante
- Department of Biostatistics, School of Public Health, Louisiana State University Health Science Center, New Orleans, Louisiana 70112
| | - Franck Mauvais-Jarvis
- the Division of Endocrinology, Tulane University School of Medicine, New Orleans, Louisiana 70112
| | - Suresh K Alahari
- From the Department of Biochemistry and Molecular Biology, School of Medicine, and
| |
Collapse
|
9
|
Ye G, Liu J, Liu Y, Chen X, Liao SF, Huang D, Huang K. Feeding glycerol-enriched yeast culture improves lactation performance, energy status, and hepatic gluconeogenic enzyme expression of dairy cows during the transition period. J Anim Sci 2016; 94:2441-50. [PMID: 27285920 DOI: 10.2527/jas.2015-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study aimed to evaluate the effects of feeding glycerol-enriched yeast culture (GY) on feed intake, lactation performance, blood metabolites, and expression of some key hepatic gluconeogenic enzymes in dairy cows during the transition period. Forty-four multiparous transition Holstein cows were blocked by parity, previous 305-d mature equivalent milk yield, and expected calving date and randomly allocated to 4 dietary treatments: Control (no additive), 2 L/d of GY (75.8 g/L glycerol and 15.3 g/L yeast), 150 g/d of glycerol (G; 0.998 g/g glycerol), and 1 L/d of yeast culture (Y; 31.1 g/L yeast). All additives were top-dressed and hand mixed into the upper one-third of the total mixed ration in the morning from -14 to +28 d relative to calving. Results indicated that the DMI, NE intake, change of BCS, and milk yields were not affected by the treatments ( > 0.05). Supplementation of GY or Y increased milk fat percentages, milk protein percentages, and milk protein yields relative to the Control or G group ( < 0.05). Cows fed GY or G had higher glucose levels and lower β-hydroxybutyric acid (BHBA) and NEFA levels in plasma than cows fed the Control ( < 0.05) and had lower NEFA levels than cows fed Y ( < 0.05). On 14 d postpartum, cows fed GY or G had higher enzyme activities, mRNA, and protein expression of cytosolic phosphoenolpyruvate carboxykinase (PEPCK-C; < 0.05); higher enzyme activities ( < 0.05) and a tendency toward higher mRNA expression ( < 0.10) of glycerol kinase (GK); and a tendency toward higher enzyme activities of pyruvate carboxylase (PC) in the liver ( < 0.10) when compared with cows fed Control or Y. The enzyme activities, mRNA, and protein expression of PEPCK-C, PC, and GK did not differ between cows fed GY and G ( > 0.10). In conclusion, dietary GY or Y supplementation increased the milk fat and protein content of the cows in early lactation and GY or G supplementation improved the energy status as indicated by greater plasma glucose and lower plasma BHBA and NEFA concentrations and upregulated the hepatic gluconeogenic enzymes of dairy cows during the transition period. Feeding cows with a GY mixture in the peripartum period combined the effects of yeast on lactation performance and the effects of glycerol on energy status in dairy cows.
Collapse
|
10
|
Seenappa V, Das B, Joshi MB, Satyamoorthy K. Context Dependent Regulation of Human Phosphoenolpyruvate Carboxykinase Isoforms by DNA Promoter Methylation and RNA Stability. J Cell Biochem 2016; 117:2506-20. [DOI: 10.1002/jcb.25543] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 03/15/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Venu Seenappa
- Department of Biotechnology; School of Life Sciences; Manipal University; Manipal India
| | - Bidyadhar Das
- Department of Zoology; Northeast Hill University; Shillong India
| | - Manjunath B. Joshi
- Department of Biotechnology; School of Life Sciences; Manipal University; Manipal India
| | - Kapaettu Satyamoorthy
- Department of Biotechnology; School of Life Sciences; Manipal University; Manipal India
| |
Collapse
|
11
|
Lelli SM, Mazzetti MB, San Martín de Viale LC. Melatonin modulates drug-induced acute porphyria. Toxicol Rep 2016; 3:141-147. [PMID: 28959532 PMCID: PMC5615779 DOI: 10.1016/j.toxrep.2015.12.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 12/29/2015] [Accepted: 12/29/2015] [Indexed: 01/10/2023] Open
Abstract
Melatonin partially eliminates the AIA/DDC-induced decrease in the activity of the gluconeogenic enzymes PEPCK and G6Pase. Melatonin favors a glucose-mediated down-regulating effect on AIA/DDC-induced ALA-S. Melatonin reduces AIA/DDC-increases in lipid peroxidation. Melatonin partially reverts the AIA/DDC-induced increase in ALA and PBG levels. The results obtained suggest the hypothetical use of Mel as co-treatment for acute porphyria.
This work investigated the modulation by melatonin (Mel) of the effects of the porphyrinogenic drugs 2-allyl-2-isopropylacetamide (AIA) and 3,5-diethoxycarbonyl-1,4-dihydro-2,4,6-collidine (DDC) on oxidative environment, glucose biosynthesis and heme pathway parameters. Administration of Mel before rat intoxication with AIA/DDC showed a clear beneficial effect in all cases. Mel induced decreases of 42% and 35% in the excretion of the hemeprecursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG), respectively, and a 33% decrease in the induction of the heme regulatory enzyme 5-aminolevulinic acid-synthase (ALA-S). The activity of the glucose metabolism enzyme phosphoenolpyruvate carboxykinase (PEPCK), which had been diminished by the porphyrinogenic treatment, was restored by 45% when animals were pre-treated with Mel. Mel abolished the modest decrease in glucose 6-phospatase (G6Pase) activity caused by AIA/DDC treatment. The oxidative status of lipids was attenuated by Mel treatment in homogenates by 47%, whereas no statistically significant AIA/DDC-induced increase in thiobarbituric acid reactive substances (TBARS) was observed in microsomes after Mel pre-treatment. We hypothesize that Mel may be scavenging reactive species of oxygen (ROS) that could be damaging lipids, PEPCK, G6Pase and ferrochelatase (FQ). Additionally, Mel administration resulted in the repression of the key enzyme ALA-S, and this could be due to an increase in glucose levels, which is known to inhibit ALA-S induction. The consequent decrease in levels of the heme precursors ALA and PBG had a beneficial effect on the drug-induced porphyria. The results obtained open the possibility of further research on the use of melatonin as a co-treatment option in acute porphyria.
Collapse
Affiliation(s)
- Sandra M Lelli
- Laboratorio de Disturbios Metabólicos por Xenobióticos, Salud Humana y Medio Ambiente, Departamento de Química Biológica, IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. II, 4to Piso, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| | - Marta B Mazzetti
- Laboratorio de Disturbios Metabólicos por Xenobióticos, Salud Humana y Medio Ambiente, Departamento de Química Biológica, IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. II, 4to Piso, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| | - Leonor C San Martín de Viale
- Laboratorio de Disturbios Metabólicos por Xenobióticos, Salud Humana y Medio Ambiente, Departamento de Química Biológica, IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Pab. II, 4to Piso, C1428EGA Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
12
|
Burak MF, Inouye KE, White A, Lee A, Tuncman G, Calay ES, Sekiya M, Tirosh A, Eguchi K, Birrane G, Lightwood D, Howells L, Odede G, Hailu H, West S, Garlish R, Neale H, Doyle C, Moore A, Hotamisligil GS. Development of a therapeutic monoclonal antibody that targets secreted fatty acid-binding protein aP2 to treat type 2 diabetes. Sci Transl Med 2015; 7:319ra205. [PMID: 26702093 DOI: 10.1126/scitranslmed.aac6336] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/04/2015] [Indexed: 09/15/2024]
Abstract
The lipid chaperone aP2/FABP4 has been implicated in the pathology of many immunometabolic diseases, including diabetes in humans, but aP2 has not yet been targeted for therapeutic applications. aP2 is not only an intracellular protein but also an active adipokine that contributes to hyperglycemia by promoting hepatic gluconeogenesis and interfering with peripheral insulin action. Serum aP2 levels are markedly elevated in mouse and human obesity and strongly correlate with metabolic complications. These observations raise the possibility of a new strategy to treat metabolic disease by targeting serum aP2 with a monoclonal antibody (mAb) to aP2. We evaluated mAbs to aP2 and identified one, CA33, that lowered fasting blood glucose, improved systemic glucose metabolism, increased systemic insulin sensitivity, and reduced fat mass and liver steatosis in obese mouse models. We examined the structure of the aP2-CA33 complex and resolved the target epitope by crystallographic studies in comparison to another mAb that lacked efficacy in vivo. In hyperinsulinemic-euglycemic clamp studies, we found that the antidiabetic effect of CA33 was predominantly linked to the regulation of hepatic glucose output and peripheral glucose utilization. The antibody had no effect in aP2-deficient mice, demonstrating its target specificity. We conclude that an aP2 mAb-mediated therapeutic constitutes a feasible approach for the treatment of diabetes.
Collapse
Affiliation(s)
- M Furkan Burak
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Karen E Inouye
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ariel White
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alexandra Lee
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Gurol Tuncman
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ediz S Calay
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Motohiro Sekiya
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Amir Tirosh
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kosei Eguchi
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | | | - Daniel Lightwood
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Louise Howells
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Geofrey Odede
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Hanna Hailu
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Shauna West
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Rachel Garlish
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Helen Neale
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Carl Doyle
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Adrian Moore
- UCB (Union Chimique Belge), 208 Bath Road, Slough, Berkshire SL1 3WE, UK
| | - Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA. Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
13
|
Winiarska K, Jarzyna R, Dzik JM, Jagielski AK, Grabowski M, Nowosielska A, Focht D, Sierakowski B. ERK1/2 pathway is involved in renal gluconeogenesis inhibition under conditions of lowered NADPH oxidase activity. Free Radic Biol Med 2015; 81:13-21. [PMID: 25601753 DOI: 10.1016/j.freeradbiomed.2014.12.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/27/2014] [Accepted: 12/26/2014] [Indexed: 01/11/2023]
Abstract
The aim of this study was to elucidate the mechanisms involved in the inhibition of renal gluconeogenesis occurring under conditions of lowered activity of NADPH oxidase (Nox), the enzyme considered to be one of the main sources of reactive oxygen species in kidneys. The in vitro experiments were performed on primary cultures of rat renal proximal tubules, with the use of apocynin, a selective Nox inhibitor, and TEMPOL (4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl), a potent superoxide radical scavenger. In the in vivo experiments, Zucker diabetic fatty (ZDF) rats, a well established model of diabetes type 2, were treated with apocynin solution in drinking water. The main in vitro findings are the following: (1) both apocynin and TEMPOL attenuate the rate of gluconeogenesis, inhibiting the step catalyzed by phosphoenolpyruvate carboxykinase (PEPCK), a key enzyme of the process; (2) in the presence of the above-noted compounds the expression of PEPCK and the phosphorylation of transcription factor CREB and ERK1/2 kinases are lowered; (3) both U0126 (MEK inhibitor) and 3-(2-aminoethyl)-5-((4-ethoxyphenyl)methylene)-2,4-thiazolidinedione (ERK inhibitor) diminish the rate of glucose synthesis via mechanisms similar to those of apocynin and TEMPOL. The observed apocynin in vivo effects include: (1) slight attenuation of hyperglycemia; (2) inhibition of renal gluconeogenesis; (3) a decrease in renal PEPCK activity and content. In view of the results summarized above, it can be concluded that: (1) the lowered activity of the ERK1/2 pathway is of importance for the inhibition of renal gluconeogenesis found under conditions of lowered superoxide radical production by Nox; (2) the mechanism of this phenomenon includes decreased PEPCK expression, resulting from diminished activity of transcription factor CREB; (3) apocynin-evoked inhibition of renal gluconeogenesis contributes to the hypoglycemic action of this compound observed in diabetic animals. Thus, the study has delivered some new insights into the recently discussed issue of the usefulness of Nox inhibition as a potential antidiabetic strategy.
Collapse
Affiliation(s)
- Katarzyna Winiarska
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Robert Jarzyna
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Jolanta M Dzik
- Department of Biochemistry, Faculty of Agriculture and Biology, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland
| | - Adam K Jagielski
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Michal Grabowski
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Agata Nowosielska
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Dorota Focht
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| | - Bartosz Sierakowski
- Department of Metabolic Regulation, Institute of Biochemistry, Faculty of Biology, University of Warsaw, I. Miecznikowa 1, 02-096 Warsaw, Poland
| |
Collapse
|
14
|
Cai D, Jia Y, Song H, Sui S, Lu J, Jiang Z, Zhao R. Betaine supplementation in maternal diet modulates the epigenetic regulation of hepatic gluconeogenic genes in neonatal piglets. PLoS One 2014; 9:e105504. [PMID: 25153319 PMCID: PMC4143294 DOI: 10.1371/journal.pone.0105504] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 07/24/2014] [Indexed: 12/31/2022] Open
Abstract
In this study, gestational sows were fed control or betaine-supplemented diets (3 g/kg) throughout the pregnancy, and the newborn piglets were used to elucidate whether maternal dietary betaine affected offspring hepatic gluconeogenic genes through epigenetic mechanisms. Neonatal piglets born to betaine-supplemented sows had significantly higher serum and hepatic betaine contents, together with significantly greater expression of methionine metabolic enzymes in the liver. Interestingly, significantly higher serum concentrations of lactic acid and glucogenic amino acids, including serine, glutamate, methionine and histidine, were detected in the piglets born to betaine-supplemented sows, which were coincident with higher hepatic glycogen content and PEPCK1 enzyme activity, as well as greater protein expression of gluconeogenic enzymes, pyruvate carboxylase (PC), cytoplasmic phosphoenolpyruvate carboxykinase (PEPCK1), mitochondrional phosphoenolpyruvate carboxykinase (PEPCK2) and fructose-1, 6-bisphosphatase (FBP1). Moreover, maternal betaine significantly changed the methylation status of both CpGs and histones on the promoter of gluconeogenic genes. The lower PEPCK1 mRNA was associated with DNA hypermethylation and more enriched repression histone mark H3K27me3, while the up-regulated PEPCK2 and FBP1 mRNA was associated with DNA hypomethylation and more enriched activation histone mark H3K4me3. Furthermore, the expression of two miRNAs predicted to target PC and 6 miRNAs predicted to target PEPCK1 was dramatically suppressed in the liver of piglets born to betaine-supplemented sows. Our results provide the first evidence that maternal betaine supplementation affects hepatic gluconeogenic genes expression in newborn piglets through enhanced hepatic methionine metabolism and epigenetic regulations, which involve DNA and histone methylations, and possibly miRNAs-mediated post-transcriptional mechanism.
Collapse
Affiliation(s)
- Demin Cai
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Yimin Jia
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Haogang Song
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Shiyan Sui
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Jingyu Lu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Zheng Jiang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
- * E-mail:
| |
Collapse
|
15
|
Massarsky A, Labarre J, Trudeau VL, Moon TW. Silver nanoparticles stimulate glycogenolysis in rainbow trout (Oncorhynchus mykiss) hepatocytes. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2014; 147:68-75. [PMID: 24374849 DOI: 10.1016/j.aquatox.2013.11.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/25/2013] [Accepted: 11/30/2013] [Indexed: 06/03/2023]
Abstract
Silver nanoparticles (AgNPs) are found in many consumer products yet their biological effects on non-target aquatic organisms are yet to be fully understood. This research aimed to investigate the effects of AgNPs on cell signaling in rainbow trout (Oncorhynchus mykiss) hepatocytes. We focused on the β-adrenoreceptor (AR), which mediates glycogenolysis, and the glucocorticoid receptor (GCR), which mediates gluconeogenesis. These two receptors have been extensively studied in trout hepatocytes due to their key roles during the stress response to increase glucose availability (among other things), allowing the organisms to cope with the stressor. We show for the first time that AgNPs at a concentration of 1 μg/mL did not interfere with the function of either the β-AR or the GCR systems in rainbow trout hepatocytes, but at the concentration of 10 μg/mL AgNPs stimulated glycogenolysis which was apparently receptor-independent. This study suggests that AgNPs could affect hormone-regulated cell signaling pathways at a concentration of 10 μg/mL.
Collapse
Affiliation(s)
- Andrey Massarsky
- Department of Biology & Centre for Advanced Research in Environmental Genomics, University of Ottawa, Ottawa, Ontario, Canada.
| | - Justine Labarre
- Department of Biology & Centre for Advanced Research in Environmental Genomics, University of Ottawa, Ottawa, Ontario, Canada
| | - Vance L Trudeau
- Department of Biology & Centre for Advanced Research in Environmental Genomics, University of Ottawa, Ottawa, Ontario, Canada
| | - Thomas W Moon
- Department of Biology & Centre for Advanced Research in Environmental Genomics, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
16
|
Kumashiro N, Beddow SA, Vatner DF, Majumdar SK, Cantley JL, Guebre-Egziabher F, Fat I, Guigni B, Jurczak MJ, Birkenfeld AL, Kahn M, Perler BK, Puchowicz MA, Manchem VP, Bhanot S, Still CD, Gerhard GS, Petersen KF, Cline GW, Shulman GI, Samuel VT. Targeting pyruvate carboxylase reduces gluconeogenesis and adiposity and improves insulin resistance. Diabetes 2013; 62:2183-94. [PMID: 23423574 PMCID: PMC3712050 DOI: 10.2337/db12-1311] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We measured the mRNA and protein expression of the key gluconeogenic enzymes in human liver biopsy specimens and found that only hepatic pyruvate carboxylase protein levels related strongly with glycemia. We assessed the role of pyruvate carboxylase in regulating glucose and lipid metabolism in rats through a loss-of-function approach using a specific antisense oligonucleotide (ASO) to decrease expression predominantly in liver and adipose tissue. Pyruvate carboxylase ASO reduced plasma glucose concentrations and the rate of endogenous glucose production in vivo. Interestingly, pyruvate carboxylase ASO also reduced adiposity, plasma lipid concentrations, and hepatic steatosis in high fat-fed rats and improved hepatic insulin sensitivity. Pyruvate carboxylase ASO had similar effects in Zucker Diabetic Fatty rats. Pyruvate carboxylase ASO did not alter de novo fatty acid synthesis, lipolysis, or hepatocyte fatty acid oxidation. In contrast, the lipid phenotype was attributed to a decrease in hepatic and adipose glycerol synthesis, which is important for fatty acid esterification when dietary fat is in excess. Tissue-specific inhibition of pyruvate carboxylase is a potential therapeutic approach for nonalcoholic fatty liver disease, hepatic insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Naoki Kumashiro
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sara A. Beddow
- Veterans Affairs Medical Center, West Haven, Connecticut
| | - Daniel F. Vatner
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sachin K. Majumdar
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer L. Cantley
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | | - Ioana Fat
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Blas Guigni
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Michael J. Jurczak
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Andreas L. Birkenfeld
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Mario Kahn
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Bryce K. Perler
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | | | - Glenn S. Gerhard
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania
| | - Kitt Falk Petersen
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Gary W. Cline
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Gerald I. Shulman
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - Varman T. Samuel
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
- Veterans Affairs Medical Center, West Haven, Connecticut
- Corresponding author: Varman T. Samuel,
| |
Collapse
|
17
|
Magnoni LJ, Scarlato NA, Patricio Ojeda F, Wöhler OC. Gluconeogenic pathway does not display metabolic cold adaptation in liver of Antarctic notothenioid fish. Polar Biol 2013. [DOI: 10.1007/s00300-013-1292-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
18
|
Maloney CA, Hay SM, Reid MD, Duncan G, Nicol F, Sinclair KD, Rees WD. A methyl-deficient diet fed to rats during the pre- and peri-conception periods of development modifies the hepatic proteome in the adult offspring. GENES AND NUTRITION 2012; 8:181-90. [PMID: 22907820 DOI: 10.1007/s12263-012-0314-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 08/02/2012] [Indexed: 01/03/2023]
Abstract
A methyl-deficient diet (MD) lacking folic acid and the associated methyl donors choline and methionine, fed to the laboratory rat during the periods of oocyte and embryo development, has been shown to programme glucose metabolism in the offspring. The hepatic proteome of the male offspring of female rats fed MD diets for 3 weeks prior to mating and for the first 5 days of gestation has been examined by 2-dimensional gel electrophoresis. Three groups of differentially abundant proteins associated with energy metabolism, amino acid metabolism and antioxidant defence were identified in the soluble proteins extracted from the liver from the MD offspring at both 6 and 12 months of age. Altered mitochondrial activity in other programming models leads to a similar pattern of differential protein abundance. Two of the differentially abundant proteins were identified as GAPDH and PGK-1 by mass spectrometry. Western blotting showed that there were multiple isoforms of both proteins with similar molecular weights but different isoelectric points. The differentially abundant spots reduced in the MD offspring corresponded to minor isoforms of GAPDH and PGK-1. The levels of PPAR-alpha, SREBP and glucocorticoid receptor mRNAs associated with other models of prenatal programming were unchanged in the MD offspring. The data suggest that a diet deficient in folic acid and associated methyl donors fed during the peri-conception and early preimplantation periods of mammalian development affects mitochondrial function in the offspring and that the posttranslational modification of proteins may be important.
Collapse
Affiliation(s)
- Christopher A Maloney
- School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | | | | | | | | | | | | |
Collapse
|
19
|
Hein GJ, Chicco A, Lombardo YB. Fish oil normalizes plasma glucose levels and improves liver carbohydrate metabolism in rats fed a sucrose-rich diet. Lipids 2011; 47:141-50. [PMID: 22045300 DOI: 10.1007/s11745-011-3623-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 10/04/2011] [Indexed: 12/23/2022]
Abstract
A sucrose-rich diet (SRD) induces insulin resistance and dyslipidemia with impaired hepatic glucose production and gluconeogenesis, accompanied by altered post-receptor insulin signaling steps. The aim of this study was to examine the effectiveness of fish oil (FO) to reverse or improve the impaired hepatic glucose metabolism once installed in rats fed 8 months a SRD. In the liver of rats fed SRD in which FO replaced corn-oil during the last 2 months, as dietary fat, several key enzyme activities and metabolites involved in glucose metabolisms (phosphorylation, glycolysis, gluconeogenesis and oxidative and non oxidative glucose pathway) were measured. The protein mass levels of IRS-1 and αp85 PI-3K at basal conditions were also analyzed. FO improved the altered activities of some enzymes involved in the glycolytic and oxidative pathways observed in the liver of SRD fed rats but was unable to restore the impaired capacity of glucose phosphorylation. Moreover, FO reversed the increase in PEPCK and G-6-Pase and reduced the G-6-Pase/GK ratio. Glycogen concentration and GSa activity returned to levels similar to those observed in the liver of the control-fed rats. Besides, FO did not modify the altered protein mass levels of IRS-1 and αp85 PI-3K. Finally, dietary FO was effective in reversing or improving the impaired activities of several key enzymes of hepatic carbohydrate metabolism contributing, at least in part, to the normalization of plasma glucose levels in the SRD-fed rats. However, these positive effects of FO were not observed under basal conditions in the early steps of insulin signaling transduction.
Collapse
Affiliation(s)
- Gustavo J Hein
- Department of Biochemistry, School of Biochemistry, University of Litoral, Ciudad Universitaria Paraje El Pozo. CC 242 (3000), Santa Fe, Argentina
| | | | | |
Collapse
|
20
|
Lu Y, Zhang G, Shen C, Uygun K, Yarmush ML, Meng Q. A novel 3D liver organoid system for elucidation of hepatic glucose metabolism. Biotechnol Bioeng 2011; 109:595-604. [PMID: 22006574 DOI: 10.1002/bit.23349] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 09/26/2011] [Accepted: 09/29/2011] [Indexed: 12/12/2022]
Abstract
Hepatic glucose metabolism is a key player in diseases such as obesity and diabetes as well as in antihyperglycemic drugs screening. Hepatocytes culture in two-dimensional configurations is limited in vitro model for hepatocytes to function properly, while truly practical platforms to perform three-dimensional (3D) culture are unavailable. In this work, we present a practical organoid culture method of hepatocytes for elucidation of glucose metabolism under nominal and stress conditions. Employing this new method of culturing cells within a hollow fiber reactor, hepatocytes were observed to self-assemble into 3D spherical organoids with preservation of tight junctions and display increased liver-specific functions. Compared to both monolayer culture and sandwich culture, the hepatocyte organoids displayed higher intracellular glycogen content, glucose consumption, and gluconeogenesis and approached the in vivo values, as also confirmed by gene expression of key enzymes. Moreover, hepatocyte organoids demonstrated more realistic sensitivity to hormonal challenges with insulin, glucagon, and dexamethasone. Finally, the exposure to high glucose demonstrated toxicities including alteration of mitochondrial membrane potential, lipid accumulation, and reactive oxygen species formation, similar to the in vivo responses, which was not captured by monolayer cultures. Collectively, hepatocyte organoids mimicked the in vivo functions better than hepatocyte monolayer and sandwich cultures, suggesting suitability for applications such as antihyperglycemic drugs screening.
Collapse
Affiliation(s)
- Yanhua Lu
- Department of Chemical and Biological Engineering, Zhejiang University, 38 Zheda Road, Hangzhou, Zhejiang 310027, China
| | | | | | | | | | | |
Collapse
|
21
|
Iwashita K, Nagashima H. Rubratoxin B induces signs of fatty acid oxidation disorders (FAODs) in mice. Toxicol Lett 2011; 206:238-43. [DOI: 10.1016/j.toxlet.2011.07.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/25/2011] [Accepted: 07/25/2011] [Indexed: 12/18/2022]
|
22
|
Cellular mechanism of insulin resistance in nonalcoholic fatty liver disease. Proc Natl Acad Sci U S A 2011; 108:16381-5. [PMID: 21930939 DOI: 10.1073/pnas.1113359108] [Citation(s) in RCA: 423] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance is associated with nonalcoholic fatty liver disease (NAFLD) and is a major factor in the pathogenesis of type 2 diabetes. The development of hepatic insulin resistance has been ascribed to multiple causes, including inflammation, endoplasmic reticulum (ER) stress, and accumulation of hepatocellular lipids in animal models of NAFLD. However, it is unknown whether these same cellular mechanisms link insulin resistance to hepatic steatosis in humans. To examine the cellular mechanisms that link hepatic steatosis to insulin resistance, we comprehensively assessed each of these pathways by using flash-frozen liver biopsies obtained from 37 obese, nondiabetic individuals and correlating key hepatic and plasma markers of inflammation, ER stress, and lipids with the homeostatic model assessment of insulin resistance index. We found that hepatic diacylglycerol (DAG) content in cytoplasmic lipid droplets was the best predictor of insulin resistance (R = 0.80, P < 0.001), and it was responsible for 64% of the variability in insulin sensitivity. Hepatic DAG content was also strongly correlated with activation of hepatic PKCε (R = 0.67, P < 0.001), which impairs insulin signaling. In contrast, there was no significant association between insulin resistance and other putative lipid metabolites or plasma or hepatic markers of inflammation. ER stress markers were only partly correlated with insulin resistance. In conclusion, these data show that hepatic DAG content in lipid droplets is the best predictor of insulin resistance in humans, and they support the hypothesis that NAFLD-associated hepatic insulin resistance is caused by an increase in hepatic DAG content, which results in activation of PKCε.
Collapse
|
23
|
How porphyrinogenic drugs modeling acute porphyria impair the hormonal status that regulates glucose metabolism. Their relevance in the onset of this disease. Toxicology 2011; 290:22-30. [PMID: 21889565 DOI: 10.1016/j.tox.2011.08.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/02/2011] [Accepted: 08/11/2011] [Indexed: 11/21/2022]
Abstract
This work deals with the study of how porphyrinogenic drugs modeling acute porphyrias interfere with the status of carbohydrate-regulating hormones in relation to key glucose enzymes and to porphyria, considering that glucose modulates the development of the disease. Female Wistar rats were treated with 2-allyl-2-isopropylacetamide (AIA) and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) using different doses of AIA (100, 250 and 500mg/kg body weight) and a single dose of DDC (50mg DDC/kg body weight). Rats were sacrificed 16h after AIA/DDC administration. In the group treated with the highest dose of AIA (group H), hepatic 5-aminolevulinic acid synthase (ALA-S) increased more than 300%, phosphoenolpyruvate carboxykinase (PEPCK) and glycogen phosphorylase (GP) activities were 43% and 46% lower than the controls, respectively, plasmatic insulin levels exceeded normal values by 617%, and plasmatic glucocorticoids (GC) decreased 20%. GC results are related to a decrease in corticosterone (CORT) adrenal production (33%) and a significant reduction in its metabolization by UDP-glucuronosyltransferase (UGT) (62%). Adrenocorticotropic hormone (ACTH) stimulated adrenal production 3-fold and drugs did not alter this process. Thus, porphyria-inducing drugs AIA and DDC dramatically altered the status of hormones that regulate carbohydrate metabolism increasing insulin levels and reducing GC production, metabolization and plasmatic levels. In this acute porphyria model, gluconeogenic and glycogenolytic blockages caused by PEPCK and GP depressed activities, respectively, would be mainly a consequence of the negative regulatory action of insulin on these enzymes. GC could also contribute to PEPCK blockage both because they were depressed by the treatment and because they are positive effectors on PEPCK. These disturbances in carbohydrates and their regulation, through ALA-S de-repression, would enhance the porphyria state promoted by the drugs on heme synthesis and destruction. This might be the mechanism underlying the "glucose effect" observed in hepatic porphyrias. The statistical correlation study performed showed association between all the variables studied and reinforce these conclusions.
Collapse
|
24
|
Potter JJ, Koteish A, Hamilton J, Liu X, Liu K, Agre P, Mezey E. Effects of acetaldehyde on hepatocyte glycerol uptake and cell size: implication of aquaporin 9. Alcohol Clin Exp Res 2011; 35:939-45. [PMID: 21294757 DOI: 10.1111/j.1530-0277.2010.01424.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The effects of ethanol and acetaldehyde on uptake of glycerol and on cell size of hepatocytes and a role Aquaporin 9 (AQP9), a glycerol transport channel, were evaluated. METHODS The studies were done in primary rat and mouse hepatocytes. The uptake of [(14) C] glycerol was determined with hepatocytes in suspension. For determination of cell size, rat hepatocytes on coated dishes were incubated with a lipophilic fluorochrome that is incorporated into the cell membrane and examined by confocal microscopy. A three-dimensional z scan of the cell was performed, and the middle slice of the z scan was used for area measurements. RESULTS Acute exposure to acetaldehyde, but not to ethanol, causes a rapid increase in the uptake of glycerol and an increase in hepatocyte size, which was inhibited by HgCl(2) , an inhibitor of aquaporins. This was not observed in hepatocytes from AQP9 knockout mice, nor observed by direct application of acetaldehyde to AQP9 expressed in Xenopus Laevis oocytes. Prolonged 24-hour exposure to either acetaldehyde or ethanol did not result in an increase in glycerol uptake by rat hepatocytes. Acetaldehyde decreased AQP9 mRNA and AQP9 protein, while ethanol decreased AQP9 mRNA but not AQP9 protein. Ethanol, but not acetaldehyde, increased the activities of glycerol kinase and phosphoenolpyruvate carboxykinase. CONCLUSIONS The acute effects of acetaldehyde, while mediated by AQP9, are probably influenced by binding of acetaldehyde to hepatocyte membranes and changes in cell permeability. The effects of ethanol in enhancing glucose kinase, and phosphoenolpyruvate carboxykinase leading to increased formation of glycerol-3-phosphate most likely contribute to alcoholic fatty liver.
Collapse
Affiliation(s)
- James J Potter
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2195, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Josefsen K, Nielsen SM, Campos A, Seifert T, Hasholt L, Nielsen JE, Nørremølle A, Skotte NH, Secher NH, Quistorff B. Reduced gluconeogenesis and lactate clearance in Huntington's disease. Neurobiol Dis 2010; 40:656-62. [DOI: 10.1016/j.nbd.2010.08.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 07/23/2010] [Accepted: 08/11/2010] [Indexed: 12/26/2022] Open
|
26
|
Hepatic 11 beta-hydroxysteroid dehydrogenase 1 involvement in alterations of glucose metabolism produced by acidotic stress in rat. J Physiol Biochem 2010; 65:329-37. [PMID: 20358345 DOI: 10.1007/bf03185927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Indexed: 10/19/2022]
Abstract
11 beta-hydroxysteroid dehydrogenase (HSDs) enzymes regulate the activity of glucocorticoids in target organs. HSD1, one of the two existing isoforms, locates mainly in CNS, liver and adipose tissue. HSD1 is involved in the pathogenesis of diseases such as obesity, insulin resistance, arterial hypertension and the Metabolic Syndrome. The stress produced by HCl overload triggers metabolic acidosis and increases liver HSD1 activity associated with increased phosphoenolpyruvate carboxykinase, a regulatory enzyme of gluconeogenesis that is activated by glucocorticoids, with increased glycaemia and glycogen breakdown. The aim of this study was to analyze whether the metabolic modifications triggered by HCl stress are due to increased liver HSD1 activity. Glycyrrhetinic acid, a potent HDS inhibitor, was administered subcutaneously (20 mg/ml) to stressed and unstressed four months old maleSprague Dawley rats to investigate changes in liver HSD1, phosphoenolpyruvate carboxykinase (PECPK) and glycogen phosphorylase activities and plasma glucose levels. It was observed that all these parameters increased in stressed animals, but that treatment with glycyrrhetinic acid significantly reduced their levels. In conclusion, our results demonstrate the involvement of HSD1 in stress induced carbohydrate disturbances and could contribute to the impact of HSD1 inhibitors on carbohydrate metabolism and its relevance in the study of Metabolic Syndrome Disorder and non insulin-dependent diabetes mellitus.
Collapse
|
27
|
Schmutz I, Ripperger JA, Baeriswyl-Aebischer S, Albrecht U. The mammalian clock component PERIOD2 coordinates circadian output by interaction with nuclear receptors. Genes Dev 2010; 24:345-57. [PMID: 20159955 DOI: 10.1101/gad.564110] [Citation(s) in RCA: 272] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Mammalian circadian clocks provide a temporal framework to synchronize biological functions. To obtain robust rhythms with a periodicity of about a day, these clocks use molecular oscillators consisting of two interlocked feedback loops. The core loop generates rhythms by transcriptional repression via the Period (PER) and Cryptochrome (CRY) proteins, whereas the stabilizing loop establishes roughly antiphasic rhythms via nuclear receptors. Nuclear receptors also govern many pathways that affect metabolism and physiology. Here we show that the core loop component PER2 can coordinate circadian output with the circadian oscillator. PER2 interacts with nuclear receptors including PPARalpha and REV-ERBalpha and serves as a coregulator of nuclear receptor-mediated transcription. Consequently, PER2 is rhythmically bound at the promoters of nuclear receptor target genes in vivo. In this way, the circadian oscillator can modulate the expression of nuclear receptor target genes like Bmal1, Hnf1alpha, and Glucose-6-phosphatase. The concept that PER2 may propagate clock information to metabolic pathways via nuclear receptors adds an important facet to the clock-dependent regulation of biological networks.
Collapse
Affiliation(s)
- Isabelle Schmutz
- Department of Medicine, Unit of Biochemistry, University of Fribourg, 1700 Fribourg, Switzerland
| | | | | | | |
Collapse
|
28
|
Stark R, Pasquel F, Turcu A, Pongratz RL, Roden M, Cline GW, Shulman GI, Kibbey RG. Phosphoenolpyruvate cycling via mitochondrial phosphoenolpyruvate carboxykinase links anaplerosis and mitochondrial GTP with insulin secretion. J Biol Chem 2009; 284:26578-90. [PMID: 19635791 PMCID: PMC2785346 DOI: 10.1074/jbc.m109.011775] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 07/15/2009] [Indexed: 11/13/2022] Open
Abstract
Pancreatic beta-cells couple the oxidation of glucose to the secretion of insulin. Apart from the canonical K(ATP)-dependent glucose-stimulated insulin secretion (GSIS), there are important K(ATP)-independent mechanisms involving both anaplerosis and mitochondrial GTP (mtGTP). How mtGTP that is trapped within the mitochondrial matrix regulates the cytosolic calcium increases that drive GSIS remains a mystery. Here we have investigated whether the mitochondrial isoform of phosphoenolpyruvate carboxykinase (PEPCK-M) is the GTPase linking hydrolysis of mtGTP made by succinyl-CoA synthetase (SCS-GTP) to an anaplerotic pathway producing phosphoenolpyruvate (PEP). Although cytosolic PEPCK (PEPCK-C) is absent, PEPCK-M message and protein were detected in INS-1 832/13 cells, rat islets, and mouse islets. PEPCK enzymatic activity is half that of primary hepatocytes and is localized exclusively to the mitochondria. Novel (13)C-labeling strategies in INS-1 832/13 cells and islets measured substantial contribution of PEPCK-M to the synthesis of PEP. As high as 30% of PEP in INS-1 832/13 cells and 41% of PEP in rat islets came from PEPCK-M. The contribution of PEPCK-M to overall PEP synthesis more than tripled with glucose stimulation. Silencing the PEPCK-M gene completely inhibited GSIS underscoring its central role in mitochondrial metabolism-mediated insulin secretion. Given that mtGTP synthesized by SCS-GTP is an indicator of TCA flux that is crucial for GSIS, PEPCK-M is a strong candidate to link mtGTP synthesis with insulin release through anaplerotic PEP cycling.
Collapse
Affiliation(s)
| | | | - Adina Turcu
- From the Departments of Internal Medicine and
| | | | - Michael Roden
- the Institute for Clinical Diabetology, German Diabetes Center, 40225 Düsseldorf, Germany
| | | | - Gerald I. Shulman
- From the Departments of Internal Medicine and
- Cellular and Molecular Physiology and
- the Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520 and
| | - Richard G. Kibbey
- From the Departments of Internal Medicine and
- Cellular and Molecular Physiology and
| |
Collapse
|
29
|
Shahsavari R, Ehsani-Zonouz A, Houshmand M, Salehnia A, Ahangari G, Firoozrai M. Plasma glucose lowering effect of the wild Satureja khuzestanica Jamzad essential oil in diabetic rats: role of decreased gluconeogenesis. Pak J Biol Sci 2009; 12:140-5. [PMID: 19579934 DOI: 10.3923/pjbs.2009.140.145] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study was to evaluate the effect of the wild SKEO on activities and genes expression of hepatic Glycogen Phosphorylase (GP) and phosphoenolpyruvate carboxykinase (PEPCK) in normal and diabetic rats. The wild SKEO was orally administered at different doses (50 and 100 mg/kg/day) to normal as well as diabetic rats for 21 days. The levels of mRNA were determined using the quantitative real-time RT-PCR technique. The plasma glucose concentrations of diabetic rats receiving SKEO (100 mg kg(-1)) compared with diabetic control were significantly decreased. Hepatic GP activity and its mRNA levels of diabetic rats treated with SKEO moderately increased. The activity of hepatic PEPCK and its mRNA levels were significantly decreased in normal rats treated with SKEO (100 mg kg(-1)). The enhancement of PEPCK activity and its mRNA levels of diabetic treated rats with SEKO (100 mg kg(-1)) was significantly decreased compared with diabetic control. In conclusion, an excessive inhibition of PEPCK in liver of diabetic rats treated with the wild SKEO may contribute to the plasma glucose lowering action of SKEO that seems to be in relation with antioxidant properties of SKEO.
Collapse
Affiliation(s)
- R Shahsavari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
30
|
Lelli SM, Mazzetti MB, San Martín de Viale LC. Hepatic alteration of tryptophan metabolism in an acute porphyria model. Biochem Pharmacol 2008; 75:704-12. [DOI: 10.1016/j.bcp.2007.09.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 09/25/2007] [Accepted: 09/27/2007] [Indexed: 10/22/2022]
|
31
|
Lavery GG, Hauton D, Hewitt KN, Brice SM, Sherlock M, Walker EA, Stewart PM. Hypoglycemia with enhanced hepatic glycogen synthesis in recombinant mice lacking hexose-6-phosphate dehydrogenase. Endocrinology 2007; 148:6100-6. [PMID: 17823265 DOI: 10.1210/en.2007-0963] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hexose-6-phosphate dehydrogenase (H6PDH) knockout (KO) mice have reduced generation of nicotinamide adenine dinucleotide phosphate (reduced) within the endoplasmic reticulum. As a consequence, 11beta-hydroxysteroid dehydrogenase type 1 enzyme activity switches from a reductase to a dehydrogenase leading to glucocorticoid inactivation. 11beta-Hydroxysteroid dehydrogenase type 1 has emerged as an important factor in regulating hepatic glucose output; therefore, we examined aspects of glucose homeostasis in KO mice. Compared with wild-type mice, KO mice reduced weight gain, displayed peripheral fasting hypoglycemia, improved glucose tolerance, and elevated plasma corticosterone concentrations. Plasma insulin levels in fed and fasted KO mice are normal; however, insulin and plasma glucose levels are reduced 4 h after fasted animals are refed, indicating improved insulin sensitivity. There is preserved induction and activity of the glucocorticoid-responsive gluconeogenic enzymes phosphoenolpyruvate carboxykinase and glucose-6-phosphatase in fasted KO mice. Glycogen storage is elevated in fed KO liver, with fed glycogenesis rates increased in KO mice. There is normal flux of lactate through gluconeogenesis recovered as plasma glucose, coupled with increased glycogen derived from lactate. These data suggest partial retention of glucocorticoid sensitivity at the level of the liver. We therefore postulate that increased glycogen synthesis may reflect increased flux of glucose-6-phosphate (H6PDH substrate) through to glycogen in the absence of H6PDH mediated metabolism.
Collapse
Affiliation(s)
- Gareth G Lavery
- Division of Medical Sciences, University of Birmingham, Birmingham, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
32
|
Rogoff D, Ryder JW, Black K, Yan Z, Burgess SC, McMillan DR, White PC. Abnormalities of glucose homeostasis and the hypothalamic-pituitary-adrenal axis in mice lacking hexose-6-phosphate dehydrogenase. Endocrinology 2007; 148:5072-80. [PMID: 17656460 DOI: 10.1210/en.2007-0593] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hexose-6-phosphate dehydrogenase (EC 1.1.1.47) catalyzes the conversion of glucose 6-phosphate to 6-phosphogluconolactone within the lumen of the endoplasmic reticulum, thereby generating reduced nicotinamide adenine dinucleotide phosphate. Reduced nicotinamide adenine dinucleotide phosphate is a necessary cofactor for the reductase activity of 11beta-hydroxysteroid dehydrogenase type 1 (EC 1.1.1.146), which converts hormonally inactive cortisone to active cortisol (in rodents, 11-dehydrocorticosterone to corticosterone). Mice with targeted inactivation of hexose-6-phosphate dehydrogenase lack 11beta-hydroxysteroid dehydrogenase type 1 reductase activity, whereas dehydrogenase activity (corticosterone to 11-dehydrocorticosterone) is increased. We now report that both glucose output and glucose use are abnormal in these mice. Mutant mice have fasting hypoglycemia. In mutant primary hepatocytes, glucose output does not increase normally in response to glucagon. Mutant animals have lower hepatic glycogen content when fed and cannot mobilize it normally when fasting. As assessed by RT-PCR, responses of hepatic enzymes to fasting are blunted; enzymes involved in gluconeogenesis (phosphoenolpyruvate carboxykinase, tyrosine aminotransferase) are not appropriately up-regulated, and expression of glucokinase, an enzyme required for glycolysis, is not suppressed. Corticosterone has attenuated effects on expression of these enzymes in cultured mutant primary hepatocytes. Mutant mice have increased sensitivity to insulin, as assessed by homeostatic model assessment values and by increased glucose uptake by the muscle. The hypothalamic-pituitary-adrenal axis is also abnormal. Circulating ACTH, deoxycorticosterone, and corticosterone levels are increased in mutant animals, suggesting decreased negative feedback on the hypothalamic-pituitary-adrenal axis. Comparison with other animal models of adrenal insufficiency suggests that many of the observed abnormalities can be explained by blunted intracellular corticosterone actions, despite elevated circulating levels of this hormone.
Collapse
Affiliation(s)
- Daniela Rogoff
- University of Texas Southwestern Medical Center, 5223 Harry Hines Boulevard, Dallas, TX 75390-9063, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
de Vries A, Holmes MC, Heijnis A, Seier JV, Heerden J, Louw J, Wolfe-Coote S, Meaney MJ, Levitt NS, Seckl JR. Prenatal dexamethasone exposure induces changes in nonhuman primate offspring cardiometabolic and hypothalamic-pituitary-adrenal axis function. J Clin Invest 2007; 117:1058-67. [PMID: 17380204 PMCID: PMC1821070 DOI: 10.1172/jci30982] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2006] [Accepted: 01/30/2007] [Indexed: 01/08/2023] Open
Abstract
Prenatal stress or glucocorticoid administration has persisting "programming" effects on offspring in rodents and other model species. Multiple doses of glucocorticoids are in widespread use in obstetric practice. To examine the clinical relevance of glucocorticoid programming, we gave 50, 120, or 200 microg/kg/d of dexamethasone (dex50, dex120, or dex200) orally from mid-term to a singleton-bearing nonhuman primate, Chlorocebus aethiops (African vervet). Dexamethasone dose-dependently reduced maternal cortisol levels without effecting maternal blood pressure, glucose, electrolytes, or weight gain. Birth weight was unaffected by any dexamethasone dose, although postnatal growth was attenuated after dex120 and dex200. At 8 months of age, dex120 and dex200 offspring showed impaired glucose tolerance and hyperinsulinemia, with reduced (approximately 25%) pancreatic beta cell number at 12 months. Dex120 and dex200 offspring had increased systolic and diastolic blood pressures at 12 months. Mild stress produced an exaggerated cortisol response in dex200 offspring, implying hypothalamic-pituitary-adrenal axis programming. The data are compatible with the extrapolation of the glucocorticoid programming hypothesis to primates and indicate that repeated glucocorticoid therapy and perhaps chronic stress in humans may have long-term effects.
Collapse
Affiliation(s)
- Annick de Vries
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Megan C. Holmes
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Areke Heijnis
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jürgen V. Seier
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Joritha Heerden
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Johan Louw
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sonia Wolfe-Coote
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Michael J. Meaney
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Naomi S. Levitt
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jonathan R. Seckl
- Endocrinology Unit, University of Edinburgh, The Queen’s Medical Research Institute, Edinburgh, United Kingdom.
Primate Unit, Diabetes Research Group, South African Medical Research Council, Cape Town, South Africa.
Douglas Hospital Research Center, McGill University, Montreal, Quebec, Canada.
Endocrine and Diabetes Unit, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
34
|
Altuna ME, Lelli SM, San Martín de Viale LC, Damasco MC. Effect of stress on hepatic 11beta-hydroxysteroid dehydrogenase activity and its influence on carbohydrate metabolism. Can J Physiol Pharmacol 2007; 84:977-84. [PMID: 17218963 DOI: 10.1139/y06-046] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stress activates the synthesis and secretion of catecholamines and adrenal glucocorticoids, increasing their circulating levels. In vivo, hepatic 11beta-hydroxysteroid dehydrogenase 1 (HSD1) stimulates the shift of 11-dehydrocorticosterone to corticosterone, enhancing active glucocorticoids at tissue level. We studied the effect of 3 types of stress, 1 induced by bucogastric overload with 200 mmol/L HCl causing metabolic acidosis (HCl), the second induced by bucogastric overload with 0.45% NaCl (NaCl), and the third induced by simulated overload (cannula), on the kinetics of hepatic HSD1 of rats and their influence on the activity of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase, glycemia, and glycogen deposition. Compared with unstressed controls, all types of stress significantly increased HSD1 activity (146% cannula, 130% NaCl, and 253% HCl), phosphoenolpyruvate carboxykinase activity (51% cannula, 48% NaCl, and 86% HCl), and glycemia (29% cannula, 30% NaCl, and 41% HCl), but decreased hepatic glycogen (68% cannula, 68% NaCl, and 78% HCl). Owing to these results, we suggest the following events occur when stress is induced: an increase in hepatic HSD1 activity, augmented active glucocorticoid levels, increased gluconeogenesis, and glycemia. Also involved are the multiple events indirectly related to glucocorticoids, which lead to the depletion of hepatic glycogen deposits, thereby contributing to increased glycemia. This new approach shows that stress increments the activity of hepatic HSD1 and suggests that this enzyme could be involved in the development of the Metabolic Syndrome.
Collapse
Affiliation(s)
- María Eugenia Altuna
- Laboratorio de Esteroides, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Departamento de Química Biológica Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, C1428EGA, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
35
|
Bernal-Mizrachi C, Xiaozhong L, Yin L, Knutsen RH, Howard MJ, Arends JJA, DeSantis P, Coleman T, Semenkovich CF. An afferent vagal nerve pathway links hepatic PPARalpha activation to glucocorticoid-induced insulin resistance and hypertension. Cell Metab 2007; 5:91-102. [PMID: 17276352 PMCID: PMC1899170 DOI: 10.1016/j.cmet.2006.12.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 08/23/2006] [Accepted: 12/20/2006] [Indexed: 11/19/2022]
Abstract
Glucocorticoid excess causes insulin resistance and hypertension. Hepatic expression of PPARalpha (Ppara) is required for glucocorticoid-induced insulin resistance. Here we demonstrate that afferent fibers of the vagus nerve interface with hepatic Ppara expression to disrupt blood pressure and glucose homeostasis in response to glucocorticoids. Selective hepatic vagotomy decreased hyperglycemia, hyperinsulinemia, hepatic insulin resistance, Ppara expression, and phosphoenolpyruvate carboxykinase (PEPCK) enzyme activity in dexamethasone-treated Ppara(+/+) mice. Selective vagotomy also decreased blood pressure, adrenergic tone, renin activity, and urinary sodium retention in these mice. Hepatic reconstitution of Ppara in nondiabetic, normotensive dexamethasone-treated PPARalpha null mice increased glucose, insulin, hepatic PEPCK enzyme activity, blood pressure, and renin activity in sham-operated animals but not hepatic-vagotomized animals. Disruption of vagal afferent fibers by chemical or surgical means prevented glucocorticoid-induced metabolic derangements. We conclude that a dynamic interaction between hepatic Ppara expression and a vagal afferent pathway is essential for glucocorticoid induction of diabetes and hypertension.
Collapse
Affiliation(s)
- Carlos Bernal-Mizrachi
- Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Liu Xiaozhong
- Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Li Yin
- Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Russell H. Knutsen
- Department of Cell Biology and Physiology, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Michael J. Howard
- Department of Neurology, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Joop J. A. Arends
- Department of Neurology, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Pascual DeSantis
- Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Trey Coleman
- Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
| | - Clay F. Semenkovich
- Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
- Department of Cell Biology and Physiology, Washington University School of Medicine, Campus Box 8127, 660 South Euclid Avenue, St. Louis, MO 63110
- *Correspondence: , Phone 314-362-4454, Fax 314-362-7641
| |
Collapse
|
36
|
Lelli SM, Ceballos NR, Mazzetti MB, Aldonatti CA, San Martín de Viale LC. Hexachlorobenzene as hormonal disruptor--studies about glucocorticoids: their hepatic receptors, adrenal synthesis and plasma levels in relation to impaired gluconeogenesis. Biochem Pharmacol 2006; 73:873-9. [PMID: 17182006 DOI: 10.1016/j.bcp.2006.11.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2006] [Revised: 11/08/2006] [Accepted: 11/14/2006] [Indexed: 11/15/2022]
Abstract
In Wistar rats, hexachlorobenzene (HCB) depresses the gluconeogenic enzyme phosphoenolpyruvate-carboxykinase (PEPCK). In the liver, glucocorticoids (GC) normally regulate the glucose synthesis by acting on PEPCK. Thus, the aim of this work was to investigate, in a time-course study, the effects of HCB on plasma GC, its adrenal synthesis and stimulation, and the kinetic parameters of its hepatic receptors (GR) in relation to the gluconeogenic blockage produced by HCB. Plasma corticosterone (CORT) concentration, urinary porphyrins and hepatic PEPCK were determined after 2, 4, 6 and 8 weeks of HCB-treatment. The effect of HCB on kinetic parameters of GR was studied in adrenalectomized porphyric rats after 2, 4 and 8 weeks of treatment. Additionally, adrenal CORT synthesis in the same weeks was measured with or without ACTH. Results show that plasma CORT in intoxicated animals dropped significantly after 2 and 4 weeks of treatment (23% and 58%, respectively), and then remained constant until the 8th week. HCB also promoted a reduction in the number of hepatic GR (50-55%) without modifying affinity. After 8 weeks, when porphyria was well established (40-50-fold increase in urinary porphyrins), a reduction (52%) in hepatic GR number, as well as a decrease in PEPCK activity (56%) were observed. Moreover, CORT biosynthesis in adrenals from intoxicated animals significantly decreased (60%) without changes in ACTH effect. Briefly, this paper shows that HCB causes a disruption in GC and GR. This disturbance could contribute to the negative effect on glucose synthesis through PEPCK regulation, thus modulating porphyria. These results enhance the knowledge about the hormonal disruption produced by chlorinated xenobiotics.
Collapse
Affiliation(s)
- Sandra M Lelli
- Laboratorio de Disturbios Metabólicos por Xenobióticos, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad Universitaria, Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
37
|
Warne JP, Horneman HF, Wick EC, Bhargava A, Pecoraro NC, Ginsberg AB, Akana SF, Dallman MF. Comparison of superior mesenteric versus jugular venous infusions of insulin in streptozotocin-diabetic rats on the choice of caloric intake, body weight, and fat stores. Endocrinology 2006; 147:5443-51. [PMID: 16873535 DOI: 10.1210/en.2006-0702] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Corticosterone (B) increases and insulin decreases food intake. However, in streptozotocin (STZ)-diabetic rats with high B, low insulin replacement promotes lard intake. To test the role of the liver on this, rats were given STZ and infused with insulin or vehicle into either the superior mesenteric or right jugular vein. Controls were nondiabetic; all rats were treated with high B. After 5 d, all rats were offered lard, 32% sucrose, chow, and water ad libitum until d 10. Diabetes exacerbated body weight loss from high B; this was prevented by insulin into the jugular, but not superior mesenteric, vein. Without insulin, STZ groups essentially consumed only chow; controls increased caloric intake about equally from the three sources. Insulin into both sites reduced chow and increased lard intake. Although circulating insulin was increased only by jugular infusion, plasma glucose and liver glycogen were similar after insulin into both sites. Fat depot weights differed: sc fat was heavier after jugular and mesenteric fat was heavier after mesenteric insulin infusions. We conclude that there are important site-specific effects of insulin in regulating the choice of, but not total, caloric intake, body weight, and fat storage in diabetic rats with high B. Furthermore, lard intake might be regulated by an insulin-derived, liver-mediated signal because superior mesenteric insulin infusion had similar effects on lard intake to jugular infusion but did not result in elevated circulating insulin levels likely associated with liver insulin removal.
Collapse
Affiliation(s)
- James P Warne
- Department of Physiology, Box 0444, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang D, Wei Y, Schmoll D, Maclean KN, Pagliassotti MJ. Endoplasmic reticulum stress increases glucose-6-phosphatase and glucose cycling in liver cells. Endocrinology 2006; 147:350-8. [PMID: 16223860 DOI: 10.1210/en.2005-1014] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Impaired regulation of hepatic glucose production is a characteristic feature of the metabolic syndrome, a cluster of diseases that includes obesity, insulin resistance, type 2 diabetes, and cardiovascular disease. It has been proposed that sustained endoplasmic reticulum stress, which appears to occur in obesity and diabetes, modulates insulin action in the liver. In this study, we show that experimental induction of endoplasmic reticulum stress increases expression and activity of glucose-6-phosphatase and the capacity for glucose release and glucose cycling in primary rat hepatocytes and H4IIE liver cells. Increased expression of the catalytic subunit of glucose-6-phosphatase was largely a result of increased transcription. Deletion analysis of the glucose-6-phosphatase promoter identified an endoplasmic reticulum stress-responsive region located between -233 and -187 with respect to the transcriptional start site. Experimental induction of endoplasmic reticulum stress increased the activity of c-jun N-terminal kinase. Prevention of endoplasmic reticulum stress-mediated activation of c-jun N-terminal kinase reduced the expression of the catalytic subunit of glucose-6-phosphatase, glucose-6-phosphatase activity, glucose release, and glucose cycling. These data demonstrate that sustained endoplasmic reticulum stress in the hepatocyte provokes adaptations, mediated in part via activation of c-jun N-terminal kinase, that act to increase hepatocellular capacity for glucose release and glucose cycling.
Collapse
Affiliation(s)
- Dong Wang
- Departments of Food Science and Human Nutrition, Colorado State University, Fort Collins, Colorado 80523-1571, USA
| | | | | | | | | |
Collapse
|
39
|
Gómez-Valadés AG, Vidal-Alabró A, Molas M, Boada J, Bermúdez J, Bartrons R, Perales JC. Overcoming diabetes-induced hyperglycemia through inhibition of hepatic phosphoenolpyruvate carboxykinase (GTP) with RNAi. Mol Ther 2005; 13:401-10. [PMID: 16271515 DOI: 10.1016/j.ymthe.2005.08.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2005] [Revised: 08/31/2005] [Accepted: 08/31/2005] [Indexed: 11/29/2022] Open
Abstract
Phosphoenolpyruvate carboxykinase (PEPCK; EC 4.1.1.32) is the rate-controlling enzyme in gluconeogenesis. In diabetic individuals, altered rates of gluconeogenesis are responsible for increased hepatic glucose output and sustained hyperglycemia. Liver-specific inhibition of PEPCK has not been assessed to date as a treatment for diabetes. We have designed a therapeutic, vector-based RNAi approach to induce posttranscriptional gene silencing of hepatic PEPCK using nonviral gene delivery. A transient reduction of PEPCK enzymatic activity (7.6 +/- 0.6 vs 9.7 +/- 1.1 mU/mg, P < 0.05) that correlated with decreased protein content of up to 50% was achieved using this strategy in diabetic mice. PEPCK partial silencing was sufficient to demonstrate lowered blood glucose (218 +/- 26 vs 364 +/- 33 mg/dl, P < 0.001) and improved glucose tolerance together with decreased circulating FFA (0.89 +/- 0.10 vs 1.44 +/- 0.11 mEq/dl, P < 0.001) and TAG (65 +/- 11 vs 102 +/- 16 mg/dl, P < 0.01), in the absence of liver steatosis or lactic acidosis. SREBP1c was down-regulated in PEPCK-silenced animals, suggesting a role for this pathway in the alterations of lipid metabolism. These data reinforce the significance of PEPCK in sustaining diabetes-induced hyperglycemia and validate liver-specific intervention at the level of PEPCK for diabetes gene therapy.
Collapse
Affiliation(s)
- Alicia G Gómez-Valadés
- Biophysics Unit, Department of Physiological Sciences II, IDIBELL-University of Barcelona, L'Hospitalet del Llobregat, Spain
| | | | | | | | | | | | | |
Collapse
|
40
|
Bizeau ME, Hazel JR. Dietary fat type alters glucose metabolism in isolated rat hepatocytes. J Nutr Biochem 2005; 10:709-15. [PMID: 15539270 DOI: 10.1016/s0955-2863(99)00060-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/1998] [Accepted: 08/18/1999] [Indexed: 10/18/2022]
Abstract
Dietary fat type can influence the regulation of carbohydrate metabolism in multiple tissue types. The influence of feeding high-fat (40% of kilocalories) diets containing either menhaden oil (MO) or coconut oil (CO) on hepatic glycogenolytic and gluconeogenic capacities was studied in isolated rat hepatocytes. Estimates of both glycogenolytic and gluconeogenic capacities were performed on hepatocytes isolated from fed and fasted animals, respectively. In MO-fed animals, both basal and hormone-stimulated rates of glucose production were significantly greater than those in CO-fed animals. However, both groups displayed a similar maximal increase in glucose production above basal for glucagon and epinephrine (2.3- and 1.9-fold, respectively). Basal rates of adenosine 3',5'-cyclic phosphate (cAMP) production were not different between groups whereas glucagon-stimulated cAMP production was increased twofold in the MO-fed group. In both MO and CO groups, the addition of 10 nM insulin reduced glucose production in fed animals to similar absolute rates. In animals fasted for 24 hours, gluconeogenic capacity was estimated using 10 mM pyruvate, lactate, or glycerol. Glucose production from all substrates was significantly greater in CO-fed animals. In addition to increased gluconeogenic rates, maximal phosphoenolpyruvate carboxykinase (PEPCK) activity was increased in the CO-fed group. Insulin reduced glucose production in both dietary groups, but the absolute rate of glucose production was 28% greater in the CO-fed group relative to the MO-fed group. In summary, dietary fat type can markedly influence the regulation of hepatic glucose metabolism in multiple metabolic pathways. MO feeding promoted glycogenolysis and sensitivity to insulin whereas CO feeding favored gluconeogenesis and reduced insulin sensitivity.
Collapse
Affiliation(s)
- M E Bizeau
- Department of Biology and The Exercise and Sport Research Institute, Arizona State University, Tempe, AZ 85287-0404, USA
| | | |
Collapse
|
41
|
Morifuji M, Sakai K, Sugiura K. Dietary whey protein modulates liver glycogen level and glycoregulatory enzyme activities in exercise-trained rats. Exp Biol Med (Maywood) 2005; 230:23-30. [PMID: 15618122 DOI: 10.1177/153537020523000103] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
This study compared the effects of dietary whey protein with dietary casein or soy protein on glycogen storage and glycoregulatory enzyme activities in the liver of sedentary and exercise-trained rats. Male Sprague-Dawley rats (ca. 130 g) were divided into one sedentary and three exercise-trained groups, with eight animals in each group. Casein was provided as the source of dietary protein in the sedentary group while the exercise-trained groups were fed casein, whey, or soy protein. Rats in the exercise-trained groups ran for 30 mins/day, 4 days/week on a motor-driven treadmill. In the exercise-trained rats, animals fed whey protein had higher liver glycogen content than animals in the other two diet groups. Glucokinase activity was significantly higher in rats fed whey protein compared to that in rats fed soy protein, while glucose 6-phosphatase activity was significantly decreased in animals on the whey protein diet compared with those the other two diets. Although 6-phospho-fructokinase activity was significantly lower in the whey protein group than in the soy protein group, we found that fructose 1,6-bisphosphatase activity was significantly higher in the whey group compared with either the casein or soy groups. Pyruvate kinase activity in rats fed the casein diet was significantly higher than in rats fed either the whey or soy protein diets. In addition, hepatic alanine aminotransferase activity and serum alanine level were also increased in the whey protein group compared with the casein or soy protein groups. Taken together, these results demonstrate that the whey protein diet in exercise-trained rats results in significantly higher levels of liver glycogen, because of the combined effects of regulation of rate limiting glycolytic and gluconeogenic enzyme activities and activation of glycogenesis from alanine via alanine amino-transferase.
Collapse
Affiliation(s)
- Masashi Morifuji
- Meiji Seika Kaisha Ltd., Health and Bioscience Laboratories, 5-3-1 Chiyoda, Sakado-shi, Saitama 350-0289, Japan.
| | | | | |
Collapse
|
42
|
Stein TP, Schluter MD, Galante AT, Soteropoulos P, Ramirez M, Bigbee A, Grindeland RE, Wade CE. Effect of hind limb muscle unloading on liver metabolism of rats. J Nutr Biochem 2005; 16:9-16. [PMID: 15629235 DOI: 10.1016/j.jnutbio.2004.07.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2004] [Revised: 06/10/2004] [Accepted: 07/09/2004] [Indexed: 12/27/2022]
Abstract
In response to decreased use, skeletal muscle undergoes an adaptive reductive remodeling. There is a shift in fiber types from slow twitch to fast twitch fiber types. Associated with muscle unloading is an increased reliance on carbohydrate metabolism for energy. The hind limb suspended (HLS) rat model was used as the experimental model to determine whether skeletal muscle unloading had any impact on the liver. We used a combination of actual enzyme assays and microarray mRNA expression to address this question. The GenMAPP program was used to identify altered metabolic pathways. We found that the major changes in the liver with HLS were increases in the expression of genes involved in the generation of energy fuels for export, specifically gluconeogenesis and lipogenesis. The expression of mRNA was increased (P<0.05) for three of the four enzymes involved in the regulation of gluconeogenesis pathway (pyruvate carboxylase (PC), phosphoenolpyruvate carboxykinase (PEPCK), and glucose-6-phosphatase (G-6-Pase). Actual assay of enzymatic activity, in micromol . min(-1) . mg protein(-1) showed G-6-Pase (0.14+0.01 vs 0.17+0.01 P<0.05), fructose 1,6, bisphophosphatase (0.048+0.002 vs 0.054+0.002, P<0.07), and PEPCK (0.031+0.002 vs 0.038+0.012 (P<0.05) to be increased. We conclude that 1) atrophied muscle is not the only tissue to be affected by HLS, as there is also a response by the liver; and 2) the major changes in liver substrate metabolism induced by HLS appear to be limited to glucose and triglyceride production. The increase in glycolytic capacity in disused muscle is paralleled by an increase in glucogenic capacity by the liver.
Collapse
Affiliation(s)
- T Peter Stein
- University of Medicine and Dentistry of New Jersey-SOM, 2 Medical Center Drive, Stratford, NJ 08084, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Taira MC, Mazzetti MB, Lelli SM, de Viale LCSM. Glycogen metabolism and glucose transport in experimental porphyria. Toxicology 2004; 197:165-75. [PMID: 15003326 DOI: 10.1016/j.tox.2003.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/30/2003] [Accepted: 12/31/2003] [Indexed: 11/26/2022]
Abstract
Hexachlorobenzene (HCB) is a fungicide of well-known porphyrinogenic ability, which induces an experimental porphyria that resembles human porphyria cutanea tarda (PCT) in several animal species. It has been demonstrated that high glucose ingestion prevents porphyria development, and high-fat/high-protein diets enhance HCB porphyrinogenic ability. On the contrary, a diet rich in carbohydrates reduces HCB effects. The aim of this work was to study HCB effects on glycogen synthesis and degradation, as well as on glucose synthesis and transport, in order to elucidate whether would justify the beneficial use of carbohydrates in this porphyria. Rats were treated with HCB dissolved in corn oil (five daily doses 100mg/kg body weight). Results showed that: (1) HCB caused an increase in glycogen content; (2) glycogen synthase activity increased three times, and phosphorylase activity decreased about 40% due to fungicide intoxication. The effect of HCB on these two activities accounted for the higher glycogen content observed in treated animals; (3) three gluconeogenic enzymes were reduced 30-50%; (4) glucose uptake in the liver decreased in all weeks studied. The alterations found in glucose synthesis, its uptake in liver and other tissues, and its release from glycogen might contribute to the biochemical porphyria picture and would account for the effect of glucose above mentioned.
Collapse
Affiliation(s)
- María Cristina Taira
- Laboratorio de Disturbios Metabólicos por Xenobióticos, Salud Humana y Medio Ambiente (DIMXSA), Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
44
|
Dunlap DY, Ikeda I, Nagashima H, Vogel CFA, Matsumura F. Effects of src-deficiency on the expression of in vivo toxicity of TCDD in a strain of c-src knockout mice procured through six generations of backcrossings to C57BL/6 mice. Toxicology 2002; 172:125-41. [PMID: 11882352 DOI: 10.1016/s0300-483x(02)00006-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The effect of TCDD was studied in c-src-deficient C57BL6-src(tm1sor) (N6 src -/- and -/+) mice, and their wild-type littermate mice (N6 src +/+). The former was created from the original strain of B6, 129-src(tm1sor) mice through six generations of backcrossings with C57BL6 mice. The results of a high dose TCDD toxicity tests in male mice indicated that N6 src-/+ mice were significantly less responsive to the toxic action of TCDD (115 microg/kg single i.p. injection) than N6 src+/+ mice in terms of reduced % body weight gain, the increase in the liver to body weight ratio, and the decrease in the adipose tissue to liver weight ratio and in the weight of pancreas. To understand the cause for these differential effects of TCDD we studied TCDD-induced changes in several biochemical parameters at day 10 and found that most drastically affected ones were glycogen depletion and phosphoenolpyruvate carboxykinase (PEPCK) downregulation. In addition, the degree of triglyceride accumulation in liver was less pronounced in N6-/+ than in N6+/+ mice. These findings suggest that the absence of c-src expression indeed affects the development of selected, TCDD-induced toxic endpoints that are related to wasting syndrome.
Collapse
Affiliation(s)
- Debra Y Dunlap
- Department of Environmental Toxicology, University of California, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
45
|
Dhahbi JM, Mote PL, Wingo J, Rowley BC, Cao SX, Walford RL, Spindler SR. Caloric restriction alters the feeding response of key metabolic enzyme genes. Mech Ageing Dev 2001; 122:1033-48. [PMID: 11389922 DOI: 10.1016/s0047-6374(01)00230-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Differential 'fuel usage' has been proposed as a mechanism for life-span extension by caloric restriction (CR). Here, we report the effects of CR, initiated after weaning, on metabolic enzyme gene expression 0, 1.5, 5, and 12 h after feeding of 24-month-old mice. Plasma glucose and insulin were reduced by approximately 20 and 80%. Therefore, apparent insulin sensitivity, as judged by the glucose to insulin ratio, increased 3.3-fold in CR mice. Phosphoenolpyruvate carboxykinase mRNA and activity were transiently reduced 1.5 h after feeding, but were 20-100% higher in CR mice at other times. Glucose-6-phosphatase mRNA was induced in CR mice and repressed in control mice before, and for 5 h following feeding. Feeding transiently induced glucokinase mRNA fourfold in control mice, but only slightly in CR mice. Pyruvate kinase and pyruvate dehydrogenase activities were reduced approximately 50% in CR mice at most times. Feeding induced glutaminase mRNA, and carbamyl phosphate synthetase I and glutamine synthase activity (and mRNA). They were each approximately twofold or higher in CR mice. These results indicate that in mice, CR maintains higher rates of gluconeogenesis and protein catabolism, even in the hours after feeding. The data are consistent with the idea that CR continuously promotes the turnover and replacement of extrahepatic proteins.
Collapse
Affiliation(s)
- J M Dhahbi
- Department of Biochemistry, University of California, Riverside, Riverside, CA 92521, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Scheuner D, Song B, McEwen E, Liu C, Laybutt R, Gillespie P, Saunders T, Bonner-Weir S, Kaufman RJ. Translational control is required for the unfolded protein response and in vivo glucose homeostasis. Mol Cell 2001; 7:1165-76. [PMID: 11430820 DOI: 10.1016/s1097-2765(01)00265-9] [Citation(s) in RCA: 1060] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The accumulation of unfolded protein in the endoplasmic reticulum (ER) attenuates protein synthesis initiation through phosphorylation of the alpha subunit of eukaryotic translation initiation factor 2 (eIF2alpha) at Ser51. Subsequently, transcription of genes encoding adaptive functions including the glucose-regulated proteins is induced. We show that eIF2alpha phosphorylation is required for translation attenuation, transcriptional induction, and survival in response to ER stress. Mice with a homozygous mutation at the eIF2alpha phosphorylation site (Ser51Ala) died within 18 hr after birth due to hypoglycemia associated with defective gluconeogenesis. In addition, homozygous mutant embryos and neonates displayed a deficiency in pancreatic beta cells. The results demonstrate that regulation of translation through eIF2alpha phosphorylation is essential for the ER stress response and in vivo glucose homeostasis.
Collapse
Affiliation(s)
- D Scheuner
- Howard Hughes Medical Institute, University of Michigan Medical Center, 48109, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Rakus D, Skalecki K, Dzugaj A. Kinetic properties of pig (Sus scrofa domestica) and bovine (Bos taurus) D-fructose-1,6-bisphosphate 1-phosphohydrolase (F1,6BPase): liver-like isozymes in mammalian lung tissue. Comp Biochem Physiol B Biochem Mol Biol 2000; 127:123-34. [PMID: 11126748 DOI: 10.1016/s0305-0491(00)00245-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
F1,6BPases from porcine and bovine lung were isolated and their kinetic properties were determined. Ks, Kis and beta were determined assuming partial-noncompetitive inhibition (simple intersecting hyperbolic noncompetitive inhibition) of the enzyme by the substrate. Values for Ks were 4.1 and 4.4 microM for porcine and bovine F1,6BPase, respectively and values for 1 were close to 0.55 in both cases. Kis were 9 and 15 microM for porcine and bovine F1,6BPase, respectively. I0.5 for AMP were determined as 7 microM for pig enzyme and 14 microM for F1,6BPase from bovine lung. The enzymes were inhibited by F2,6BP with Ki's of 0.19 and 0.21 microM for porcine and bovine enzymes, respectively. In the presence of AMP concentration equal to I0.5, the Ki values for pig and bovine enzymes were 0.07 and 0.09 microM, respectively. The levels of F2,6BP, AMP and antioxidant enzymes activities in pig and bovine lung tissues were also determined. The cDNA coding sequence of pig lung F1,6BPase1 showed a high homology with pig liver enzyme, differing only in four positions (G/C-63, T/A-808, G/C-884 and T/A-1005) resulting in a single amino acid substitution (Gly-295 for Ala-295). It is hypothesized that the lung F1,6BPase participates in gluconeogenesis, surfactant synthesis and antioxidant reactions.
Collapse
Affiliation(s)
- D Rakus
- Department of Animal Physiology, Zoological Institute, Wroclaw University, Cybulskiego, Poland
| | | | | |
Collapse
|
48
|
Sea MM, Fong WP, Huang Y, Chen ZY. Weight cycling-induced alteration in fatty acid metabolism. Am J Physiol Regul Integr Comp Physiol 2000; 279:R1145-55. [PMID: 10956277 DOI: 10.1152/ajpregu.2000.279.3.r1145] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epidemiological studies have suggested that repeated weight cycling over time may increase the risk of coronary heart disease. The mechanism involved remains poorly understood, but the change in lipid metabolism during weight cycling has been offered as a possible explanation. The present study investigated the effect of weight cycling on the size and fatty acid composition of rat fat pads as well as serum cholesterol, triglyceride, glucose, insulin, and glucagon in rats. Two consecutive weight cycles were induced by 40% energy restriction followed by ad libitum refeeding of either a moderate-fat (MF; 22% energy) or a high-fat (HF; 45% energy) diet. The lipogenic enzymes, including fatty acid synthase, acetyl-CoA carboxylase, malic enzyme, pyruvate kinase, and lipoprotein lipase in the weight-cycled (WC) rats fed only the HF diet, yielded an overshoot of activities at the end of two weight cycles. These changes were accompanied by an 80% increase in the size of the adipocyte and a 40-50% increase in the size of perirenal and epididymal fat tissues in HF-WC rats. Regardless of whether the rats were fed the HF or MF diet, all WC rats showed a gradual reduction in linoleic and alpha-linolenic acid and an increase in palmitic, palmitoleic, and stearic acid in total body lipid. It is concluded that weight cycling in rats may promote body fatness if an HF diet is consumed and can significantly alter whole body fatty acid balance irrespective of whether they consumed an MF or HF diet. Most importantly, the weight cycling led to an overshoot or fluctuation of serum cholesterol, triglyceride, glucose, insulin, and glucagon. If weight cycling is associated with an increased risk of cardiovascular disease, then, part of the mechanism may involve the changes in these risk factors.
Collapse
Affiliation(s)
- M M Sea
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | | | | | | |
Collapse
|
49
|
Cheema SK, Clandinin MT. Dietary fat-induced suppression of lipogenic enzymes in B/B rats during the development of diabetes. Lipids 2000; 35:421-5. [PMID: 10858027 DOI: 10.1007/s11745-000-540-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
This study was designed to determine the level of inhibition of gene transcription by the reduction in insulin levels upon the onset of diabetes in spontaneously diabetic B/B rats and if reducing the level of polyunsaturated fatty acids (PUFA) in the diet will increase lipogenic enzyme activity. Control (eight animals per group) and spontaneously diabetic B/B male weanling rats (25 animals per group) were fed semipurified diets containing 20% (w/w) fat of either low (0.25) or high (1.0) polyunsaturated to saturated (P/S) fatty acid ratio. Rats were killed at the onset of diabetes [blood glucose level of approximately/= 100 mg/dL (5.55 mM)] and as they became highly diabetic [blood glucose level of approximately/= 400 mg/dL (22.22 mM)]. Total RNA was extracted from liver, and the relative amount of mRNA coding for fatty acid synthase (FAS), acetyl-CoA carboxylase, malic enzyme, pyruvate kinase, and phosphoenolpyruvate carboxykinase was determined. Liver enzyme activities were also measured. The mRNA levels for FAS, acetyl-CoA carboxylase, and malic enzyme decreased compared to control animals. The mRNA level for pyruvate kinase decreased at the onset of diabetes as compared to control animals. Feeding animals the low P/S diet treatment elevated the level of mRNA and lipogenic enzyme activity compared to animals fed the high P/S diet treatment, suggesting that the effect of PUFA on lipogenic enzymes is through a direct effect on gene expression.
Collapse
Affiliation(s)
- S K Cheema
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
50
|
Dhahbi JM, Mote PL, Wingo J, Tillman JB, Walford RL, Spindler SR. Calories and aging alter gene expression for gluconeogenic, glycolytic, and nitrogen-metabolizing enzymes. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:E352-60. [PMID: 10444432 DOI: 10.1152/ajpendo.1999.277.2.e352] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We characterized the effects of calorie restriction (CR) on the expression of key glycolytic, gluconeogenic, and nitrogen-metabolizing enzymes in mice. Of the gluconeogenic enzymes investigated, liver glucose-6-phosphatase mRNA increased 1.7- and 2. 3-fold in young and old CR mice. Phosphoenolpyruvate carboxykinase mRNA and activity increased 2.5- and 1.7-fold in old CR mice. Of the key glycolytic enzymes, pyruvate kinase mRNA and activity decreased approximately 60% in CR mice. Hepatic phosphofructokinase-1 and pyruvate dehydrogenase mRNA decreased 10-20% in CR mice. Of the genes that detoxify ammonia generated from protein catabolism, hepatic glutaminase, carbamyl phosphate synthase I, and tyrosine aminotransferase mRNAs increased 2.4-, 1.8-, and 1.8-fold with CR, respectively. Muscle glutamine synthetase mRNA increased 1.3- and 2. 1-fold in young and old CR mice. Hepatic glutamine synthetase mRNA and activity each decreased 38% in CR mice. These CR-induced changes are consistent with other studies suggesting that CR may decrease enzymatic capacity for glycolysis and increase the enzymatic capacity for hepatic gluconeogenesis and the disposal of byproducts of muscle protein catabolism.
Collapse
Affiliation(s)
- J M Dhahbi
- Department of Biochemistry, University of California, Riverside 92521, California, USA
| | | | | | | | | | | |
Collapse
|