1
|
Moreno-Vargas AD, Andrade-Cetto A, Espinoza-Hernández FA, Mata-Torres G. Proposed mechanisms of action participating in the hypoglycemic effect of the traditionally used Croton guatemalensis Lotsy and junceic acid, its main compound. Front Pharmacol 2024; 15:1436927. [PMID: 39478960 PMCID: PMC11521914 DOI: 10.3389/fphar.2024.1436927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
Croton guatemalensis Lotsy (Euphorbiaceae) is an important traditional medicine that is used by the Cakchiquels of Guatemala to control hyperglycemia in patients with type 2 diabetes. Previous studies have shown that administration of this plant induces an acute hypoglycemic effect during fasting and that the main compound is junceic acid, a diterpenoid with a clerodane skeleton; however, junceic acid has not been reported to have hypoglycemic activity in the literature. As the mechanisms involved in the hypoglycemic effect of C. guatemalensis remain unknown, the objective of the present investigation was to elucidate the hypoglycemic mechanisms of this species, as well as its major compound, junceic acid. The results indicated that, similar to complete extract, junceic acid exhibited a hypoglycemic effect in hyperglycemic rats. Both C. guatemalensis extract and junceic acid inhibited the activity of two rate-limiting enzymes involved in hepatic glucose production; however, compared with chlorogenic acid, junceic acid had a more potent effect on glucose-6-phosphatase levels than chlorogenic acid, which was used as a positive control. Furthermore, both fasting and postprandial insulin levels decreased in healthy and hyperglycemic rats despite reduced blood glucose levels in both metabolic states, suggesting a potential insulin-sensitizing effect. However, neither of these compounds potentiated the effect of insulin in insulin tolerance tests nor inhibited the enzyme activity of protein tyrosine phosphatase 1B, a negative regulator of the insulin pathway. Therefore, the insulin-sensitizing effect is thought to be independent of insulin and mediated by potential activation of the AMP-activated protein kinase pathway. The specific activation of this master regulator in β-cells results in the inhibition of insulin secretion in a healthy state and the restoration of the insulin response under conditions of glucotoxicity; these effects were observed after the administration of the extract and junceic acid in healthy and hyperglycemic rats. Overall, the main findings of this study establish a basis of the mechanisms of action of C. guatemalensis and its main compound, junceic acid, in terms of their hypoglycemic effect.
Collapse
Affiliation(s)
- Angelina Daniela Moreno-Vargas
- Laboratorio de Etnofarmacología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Ciudad Universitaria, Coyoacán, Mexico
| | - Adolfo Andrade-Cetto
- Laboratorio de Etnofarmacología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Mexico
| | | | - Gerardo Mata-Torres
- Laboratorio de Etnofarmacología, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad Universitaria, Coyoacán, Mexico
| |
Collapse
|
2
|
Modeling the progression of Type 2 diabetes with underlying obesity. PLoS Comput Biol 2023; 19:e1010914. [PMID: 36848379 PMCID: PMC9997875 DOI: 10.1371/journal.pcbi.1010914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 03/09/2023] [Accepted: 02/01/2023] [Indexed: 03/01/2023] Open
Abstract
Environmentally induced or epigenetic-related beta-cell dysfunction and insulin resistance play a critical role in the progression to diabetes. We developed a mathematical modeling framework capable of studying the progression to diabetes incorporating various diabetogenic factors. Considering the heightened risk of beta-cell defects induced by obesity, we focused on the obesity-diabetes model to further investigate the influence of obesity on beta-cell function and glucose regulation. The model characterizes individualized glucose and insulin dynamics over the span of a lifetime. We then fit the model to the longitudinal data of the Pima Indian population, which captures both the fluctuations and long-term trends of glucose levels. As predicted, controlling or eradicating the obesity-related factor can alleviate, postpone, or even reverse diabetes. Furthermore, our results reveal that distinct abnormalities of beta-cell function and levels of insulin resistance among individuals contribute to different risks of diabetes. This study may encourage precise interventions to prevent diabetes and facilitate individualized patient treatment.
Collapse
|
3
|
Yang CH, Mangiafico SP, Waibel M, Loudovaris T, Loh K, Thomas HE, Morahan G, Andrikopoulos S. E2f8 and Dlg2 genes have independent effects on impaired insulin secretion associated with hyperglycaemia. Diabetologia 2020; 63:1333-1348. [PMID: 32356104 DOI: 10.1007/s00125-020-05137-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 02/14/2020] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Reduced insulin secretion results in hyperglycaemia and diabetes involving a complex aetiology that is yet to be fully elucidated. Genetic susceptibility is a key factor in beta cell dysfunction and hyperglycaemia but the responsible genes have not been defined. The Collaborative Cross (CC) is a recombinant inbred mouse panel with diverse genetic backgrounds allowing the identification of complex trait genes that are relevant to human diseases. The aim of this study was to identify and characterise genes associated with hyperglycaemia. METHODS Using an unbiased genome-wide association study, we examined random blood glucose and insulin sensitivity in 53 genetically unique mouse strains from the CC population. The influences of hyperglycaemia susceptibility quantitative trait loci (QTLs) were investigated by examining glucose tolerance, insulin secretion, pancreatic histology and gene expression in the susceptible mice. Expression of candidate genes and their association with insulin secretion were examined in human islets. Mechanisms underlying reduced insulin secretion were studied in MIN6 cells using RNA interference. RESULTS Wide variations in blood glucose levels and the related metabolic traits (insulin sensitivity and body weight) were observed in the CC population. We showed that elevated blood glucose in the CC strains was not due to insulin resistance nor obesity but resulted from reduced insulin secretion. This insulin secretory defect was demonstrated to be independent of abnormalities in islet morphology, beta cell mass and pancreatic insulin content. Gene mapping identified the E2f8 (p = 2.19 × 10-15) and Dlg2 loci (p = 3.83 × 10-8) on chromosome 7 to be significantly associated with hyperglycaemia susceptibility. Fine mapping the implicated regions using congenic mice demonstrated that these two loci have independent effects on insulin secretion in vivo. Significantly, our results revealed that increased E2F8 and DLG2 gene expression are correlated with enhanced insulin secretory function in human islets. Furthermore, loss-of-function studies in MIN6 cells demonstrated that E2f8 is involved in insulin secretion through an ATP-sensitive K+ channel-dependent pathway, which leads to a 30% reduction in Abcc8 expression. Similarly, knockdown of Dlg2 gene expression resulted in impaired insulin secretion in response to glucose and non-glucose stimuli. CONCLUSIONS/INTERPRETATION Collectively, these findings suggest that E2F transcription factor 8 (E2F8) and discs large homologue 2 (DLG2) regulate insulin secretion. The CC resource enables the identification of E2f8 and Dlg2 as novel genes associated with hyperglycaemia due to reduced insulin secretion in pancreatic beta cells. Taken together, our results provide better understanding of the molecular control of insulin secretion and further support the use of the CC resource to identify novel genes relevant to human diseases.
Collapse
Affiliation(s)
- Chieh-Hsin Yang
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Level 7, Lance Townsend Building, Studley Road, Heidelberg, VIC, 3084, Australia.
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia.
| | - Salvatore P Mangiafico
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Level 7, Lance Townsend Building, Studley Road, Heidelberg, VIC, 3084, Australia
| | - Michaela Waibel
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Thomas Loudovaris
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Kim Loh
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Helen E Thomas
- St Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, VIC, 3065, Australia
| | - Grant Morahan
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Sofianos Andrikopoulos
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Level 7, Lance Townsend Building, Studley Road, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
4
|
Nolan CJ, Prentki M. Insulin resistance and insulin hypersecretion in the metabolic syndrome and type 2 diabetes: Time for a conceptual framework shift. Diab Vasc Dis Res 2019; 16:118-127. [PMID: 30770030 DOI: 10.1177/1479164119827611] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
While few dispute the existence of the metabolic syndrome as a clustering of factors indicative of poor metabolic health, its utility above that of its individual components in the clinical care of individual patients is questioned. This is likely a consequence of the failure of clinicians and scientists to agree on a unifying mechanism to explain the metabolic syndrome. Insulin resistance has most commonly been proposed for this role and is generally considered to be a root causative factor for not only metabolic syndrome but also for its associated conditions of non-alcoholic fatty liver disease (NAFLD), polycystic ovary syndrome (PCOS), obesity-related type 2 diabetes (T2D) and atherosclerotic cardiovascular disease (ASCVD). An alternative view, for which evidence is mounting, is that hyper-responsiveness of islet β-cells to a hostile environment, such as westernised lifestyle, is primary and that the resulting hyperinsulinaemia drives the other components of the metabolic syndrome. Importantly, within this new conceptual framework, insulin resistance, while always a biomarker and state of poor metabolic health, is not considered to be harmful, but a protective adaptive response of critical tissues including the myocardium against insulin-induced metabolic stress. This major shift in how metabolic syndrome can be considered puts insulin hypersecretion into position as the unifying mechanism. If shown to be correct, this new conceptual framework has major implications for the future prevention and management of the metabolic syndrome, including its associated conditions of NAFLD, PCOS, obesity-related T2D and ASCVD.
Collapse
Affiliation(s)
- Christopher J Nolan
- 1 Department of Endocrinology, The Canberra Hospital, Garran, ACT, Australia
- 2 Australian National University Medical School and John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Marc Prentki
- 3 CRCHUM and Montreal Diabetes Research Center, University of Montreal, Montreal, QC, Canada
- 4 Department of Nutrition and Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
5
|
Lamontagne J, Al-Mass A, Nolan CJ, Corkey BE, Madiraju SRM, Joly E, Prentki M. Identification of the signals for glucose-induced insulin secretion in INS1 (832/13) β-cells using metformin-induced metabolic deceleration as a model. J Biol Chem 2017; 292:19458-19468. [PMID: 28972173 DOI: 10.1074/jbc.m117.808105] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/13/2017] [Indexed: 12/23/2022] Open
Abstract
Metabolic deceleration in pancreatic β-cells is associated with inhibition of glucose-induced insulin secretion (GIIS), but only in the presence of intermediate/submaximal glucose concentrations. Here, we used acute metformin treatment as a tool to induce metabolic deceleration in INS1 (832/13) β-cells, with the goal of identifying key pathways and metabolites involved in GIIS. Metabolites and pathways previously implicated as signals for GIIS were measured in the cells at 2-25 mm glucose, with or without 5 mm metformin. We defined three criteria to identify candidate signals: 1) glucose-responsiveness, 2) sensitivity to metformin-induced inhibition of the glucose effect at intermediate glucose concentrations, and 3) alleviation of metformin inhibition by elevated glucose concentrations. Despite the lack of recovery from metformin-induced impairment of mitochondrial energy metabolism (glucose oxidation, O2 consumption, and ATP production), insulin secretion was almost completely restored at elevated glucose concentrations. Meeting the criteria for candidates involved in promoting GIIS were the following metabolic indicators and metabolites: cytosolic NAD+/NADH ratio (inferred from the dihydroxyacetone phosphate:glycerol-3-phosphate ratio), mitochondrial membrane potential, ADP, Ca2+, 1-monoacylglycerol, diacylglycerol, malonyl-CoA, and HMG-CoA. On the contrary, most of the purine and nicotinamide nucleotides, acetoacetyl-CoA, H2O2, reduced glutathione, and 2-monoacylglycerol were not glucose-responsive. Overall these results underscore the significance of mitochondrial energy metabolism-independent signals in GIIS regulation; in particular, the candidate lipid signaling molecules 1-monoacylglycerol, diacylglycerol, and malonyl-CoA; the predominance of KATP/Ca2+ signaling control by low ADP·Mg2+ rather than by high ATP levels; and a role for a more oxidized state (NAD+/NADH) in the cytosol during GIIS that favors high glycolysis rates.
Collapse
Affiliation(s)
- Julien Lamontagne
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada
| | - Anfal Al-Mass
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada.,the Department of Medicine, McGill University, Montréal, Québec H4A 3J1, Canada
| | - Christopher J Nolan
- the Department of Endocrinology, Canberra Hospital and the Medical School, Australian National University, Canberra ACT 2605, Australia, and
| | - Barbara E Corkey
- the Department of Medicine, Obesity Research Center, Boston University School of Medicine, Boston, Massachusetts 02118
| | - S R Murthy Madiraju
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada
| | - Erik Joly
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada
| | - Marc Prentki
- From the Molecular Nutrition Unit and Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec H2X 0A9, Canada, .,the Departments of Nutrition and Biochemistry, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| |
Collapse
|
6
|
Liu C, Fu Y, Li CE, Chen T, Li X. Phycocyanin-Functionalized Selenium Nanoparticles Reverse Palmitic Acid-Induced Pancreatic β Cell Apoptosis by Enhancing Cellular Uptake and Blocking Reactive Oxygen Species (ROS)-Mediated Mitochondria Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:4405-4413. [PMID: 28510423 DOI: 10.1021/acs.jafc.7b00896] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Accumulation of palmitic acid (PA) in human bodies could cause damage to pancreatic β cells and lead to chronic diseases by generation of reactive oxygen species (ROS). Therefore, it is of great significance to search for nutrition-available agents with antioxidant activity to protect pancreatic islet cells against PA-induced damage. Phycocyanin (PC) and selenium (Se) have been reported to have excellent antioxidant activity. In this study, PC-functionalized selenium nanoparticles (PC-SeNPs) were synthesized to investigate the in vitro protective effects on INS-1E rat insulinoma β cells against PA-induced cell death. A potent protective effect was achieved by regulation of particle size and PC content. Among three PC-SeNPs (165, 235, and 371 nm), PC-SeNPs-235 nm showed the highest cellular uptake and the best protective activities. For cell cycle analysis, PC-SeNPs showed a better protective effect on PA-induced INS-1E cell apoptosis than PC or SeNPs, and PC-SeNPs-235 nm exhibited the best effect. Further mechanistic studies demonstrated that PA induced overproduction of intracellular ROS, mitochondria fragmentation, activation of caspase-3, -8, and -9, and cleavage of PARP. However, pretreatment of the cells with PC-SeNPs effectively blocked these intracellular events, which suggests that PC-SeNPs could protect INS-1E cells against PA-induced cell apoptosis via attenuating oxidative stress and downstream signaling pathways. This finding provides a great promising nutritional approach for protection against diseases related to islet damage.
Collapse
Affiliation(s)
- Chang Liu
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Yuanting Fu
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Chang-E Li
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Jinan University , Guangzhou 510632, China
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University , Guangzhou 510632, China
| |
Collapse
|
7
|
Metherell LA, Guerra-Assunção JA, Sternberg MJ, David A. Three-Dimensional Model of Human Nicotinamide Nucleotide Transhydrogenase (NNT) and Sequence-Structure Analysis of its Disease-Causing Variations. Hum Mutat 2016; 37:1074-84. [PMID: 27459240 PMCID: PMC5026163 DOI: 10.1002/humu.23046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 06/23/2016] [Accepted: 06/28/2016] [Indexed: 12/22/2022]
Abstract
Defective mitochondrial proteins are emerging as major contributors to human disease. Nicotinamide nucleotide transhydrogenase (NNT), a widely expressed mitochondrial protein, has a crucial role in the defence against oxidative stress. NNT variations have recently been reported in patients with familial glucocorticoid deficiency (FGD) and in patients with heart failure. Moreover, knockout animal models suggest that NNT has a major role in diabetes mellitus and obesity. In this study, we used experimental structures of bacterial transhydrogenases to generate a structural model of human NNT (H‐NNT). Structure‐based analysis allowed the identification of H‐NNT residues forming the NAD binding site, the proton canal and the large interaction site on the H‐NNT dimer. In addition, we were able to identify key motifs that allow conformational changes adopted by domain III in relation to its functional status, such as the flexible linker between domains II and III and the salt bridge formed by H‐NNT Arg882 and Asp830. Moreover, integration of sequence and structure data allowed us to study the structural and functional effect of deleterious amino acid substitutions causing FGD and left ventricular non‐compaction cardiomyopathy. In conclusion, interpretation of the function–structure relationship of H‐NNT contributes to our understanding of mitochondrial disorders.
Collapse
Affiliation(s)
- Louise A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - José Afonso Guerra-Assunção
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Michael J Sternberg
- Centre for Integrative System Biology and Bioinformatics, Imperial College London, London, UK
| | - Alessia David
- Centre for Integrative System Biology and Bioinformatics, Imperial College London, London, UK.
| |
Collapse
|
8
|
Pepin É, Al-Mass A, Attané C, Zhang K, Lamontagne J, Lussier R, Madiraju SRM, Joly E, Ruderman NB, Sladek R, Prentki M, Peyot ML. Pancreatic β-Cell Dysfunction in Diet-Induced Obese Mice: Roles of AMP-Kinase, Protein Kinase Cε, Mitochondrial and Cholesterol Metabolism, and Alterations in Gene Expression. PLoS One 2016; 11:e0153017. [PMID: 27043434 PMCID: PMC4820227 DOI: 10.1371/journal.pone.0153017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 03/22/2016] [Indexed: 12/27/2022] Open
Abstract
Diet induced obese (DIO) mice can be stratified according to their weight gain in response to high fat diet as low responders (LDR) and high responders (HDR). This allows the study of β-cell failure and the transitions to prediabetes (LDR) and early diabetes (HDR). C57BL/6N mice were fed for 8 weeks with a normal chow diet (ND) or a high fat diet and stratified as LDR and HDR. Freshly isolated islets from ND, LDR and HDR mice were studied ex-vivo for mitochondrial metabolism, AMPK activity and signalling, the expression and activity of key enzymes of energy metabolism, cholesterol synthesis, and mRNA profiling. Severely compromised glucose-induced insulin secretion in HDR islets, as compared to ND and LDR islets, was associated with suppressed AMP-kinase activity. HDR islets also showed reduced acetyl-CoA carboxylase activity and enhanced activity of 3-hydroxy-3-methylglutaryl-CoA reductase, which led respectively to elevated fatty acid oxidation and increased cholesterol biosynthesis. HDR islets also displayed mitochondrial membrane hyperpolarization and reduced ATP turnover in the presence of elevated glucose. Expression of protein kinase Cε, which reduces both lipolysis and production of signals for insulin secretion, was elevated in DIO islets. Genes whose expression increased or decreased by more than 1.2-fold were minor between LDR and ND islets (17 differentially expressed), but were prominent between HDR and ND islets (1508 differentially expressed). In HDR islets, particularly affected genes were related to cell cycle and proliferation, AMPK signaling, mitochondrial metabolism and cholesterol metabolism. In conclusion, chronically reduced AMPK activity, mitochondrial dysfunction, elevated cholesterol biosynthesis in islets, and substantial alterations in gene expression accompany β-cell failure in HDR islets. The β-cell compensation process in the prediabetic state (LDR) is largely independent of transcriptional adaptive changes, whereas the transition to early diabetes (HDR) is associated with major alterations in gene expression.
Collapse
Affiliation(s)
- Émilie Pepin
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Anfal Al-Mass
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Camille Attané
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Kezhuo Zhang
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Julien Lamontagne
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Roxane Lussier
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - S. R. Murthy Madiraju
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Erik Joly
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Neil B. Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine and Diabetes Unit, Boston Medical Center, Boston, MA, United States of America
| | - Robert Sladek
- Departments of Medicine and Human Genetics, McGill University, Montreal, Québec, Canada
| | - Marc Prentki
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- Departments of Nutrition, Biochemistry and Molecular Medicine, Faculty of Medicine, University of Montréal, Montreal, Québec, Canada
- * E-mail: (MP); (MLP)
| | - Marie-Line Peyot
- Montreal Diabetes Research Center and Centre de Recherche du CHUM, Montréal, Québec, Canada
- * E-mail: (MP); (MLP)
| |
Collapse
|
9
|
Lamont BJ, Waters MF, Andrikopoulos S. A low-carbohydrate high-fat diet increases weight gain and does not improve glucose tolerance, insulin secretion or β-cell mass in NZO mice. Nutr Diabetes 2016; 6:e194. [PMID: 26878317 PMCID: PMC4775822 DOI: 10.1038/nutd.2016.2] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/25/2015] [Accepted: 12/22/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/OBJECTIVES Dietary guidelines for the past 20 years have recommended that dietary fat should be minimized. In contrast, recent studies have suggested that there could be some potential benefits for reducing carbohydrate intake in favor of increased fat. It has also been suggested that low-carbohydrate diets be recommended for people with type 2 diabetes. However, whether such diets can improve glycemic control will likely depend on their ability to improve β-cell function, which has not been studied. The objective of the study was to assess whether a low-carbohydrate and therefore high-fat diet (LCHFD) is beneficial for improving the endogenous insulin secretory response to glucose in prediabetic New Zealand Obese (NZO) mice. METHODS NZO mice were maintained on either standard rodent chow or an LCHFD from 6 to 15 weeks of age. Body weight, food intake and blood glucose were assessed weekly. Blood glucose and insulin levels were also assessed after fasting and re-feeding and during an oral glucose tolerance test. The capacity of pancreatic β-cells to secrete insulin was assessed in vivo with an intravenous glucose tolerance test. β-Cell mass was assessed in histological sections of pancreata collected at the end of the study. RESULTS In NZO mice, an LCHFD reduced plasma triglycerides (P=0.001) but increased weight gain (P<0.0001), adipose tissue mass (P=0.0015), high-density lipoprotein cholesterol (P=0.044) and exacerbated glucose intolerance (P=0.013). Although fasting insulin levels tended to be higher (P=0.08), insulin secretory function in LCHFD-fed mice was not improved (P=0.93) nor was β-cell mass (P=0.75). CONCLUSIONS An LCHFD is unlikely to be of benefit for preventing the decline in β-cell function associated with the progression of hyperglycemia in type 2 diabetes.
Collapse
Affiliation(s)
- B J Lamont
- Department of Medicine, Austin Hospital, The University of Melbourne, Heidelberg, Victoria, Australia
| | - M F Waters
- Department of Medicine, Austin Hospital, The University of Melbourne, Heidelberg, Victoria, Australia
| | - S Andrikopoulos
- Department of Medicine, Austin Hospital, The University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
10
|
Lombardo GE, Arcidiacono B, De Rose RF, Lepore SM, Costa N, Montalcini T, Brunetti A, Russo D, De Sarro G, Celano M. Normocaloric Diet Restores Weight Gain and Insulin Sensitivity in Obese Mice. Front Endocrinol (Lausanne) 2016; 7:49. [PMID: 27303363 PMCID: PMC4882321 DOI: 10.3389/fendo.2016.00049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/09/2016] [Indexed: 12/17/2022] Open
Abstract
An increased incidence of obesity is registered worldwide, and its association with insulin resistance and type 2 diabetes is closely related with increased morbidity and mortality for cardiovascular diseases. A major clinical problem in the management of obesity is the non-adherence or low adherence of patients to a hypocaloric dietetic restriction. In this study, we evaluated in obese mice the effects of shifting from high-calorie foods to normal diet on insulin sensitivity. Male C57BL/6JOlaHsd mice (n = 20) were fed with high fat diet (HFD) for a 24-week period. Afterward, body weight, energy, and food intake were measured in all animals, together with parameters of insulin sensitivity by homeostatic model assessment of insulin resistance and plasma glucose levels in response to insulin administration. Moreover, in half of these mice, Glut4 mRNA levels were measured in muscle at the end of the high fat treatment, whereas the rest of the animals (n = 10) were shifted to normocaloric diet (NCD) for 10 weeks, after which the same analyses were carried out. A significant reduction of body weight was found after the transition from high to normal fat diet, and this decrease correlated well with an improvement in insulin sensitivity. In fact, we found a reduction in serum insulin levels and the recovery of insulin responsiveness in terms of glucose disposal measured by insulin tolerance test and Glut4 mRNA and protein expression. These results indicate that obesity-related insulin resistance may be rescued by shifting from HFD to NCD.
Collapse
Affiliation(s)
| | - Biagio Arcidiacono
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | | | - Saverio Massimo Lepore
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Nicola Costa
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti, ; Diego Russo,
| | - Diego Russo
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- *Correspondence: Antonio Brunetti, ; Diego Russo,
| | | | - Marilena Celano
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
11
|
Abstract
Type 2 diabetes (T2D) represents a significant global epidemic with more than 285 million people affected worldwide. Regulating glycemia in T2D patients can be partially achieved with currently available treatment, but intensive research during the last decades have led to the discovery of modified compounds or new targets that could represent great hope for safe and effective treatment in the future. Among them, targets in the CNS that are known to control feeding and body weight have been also shown to exert glucoregulatory actions, and could be a key in the development of a new generation of drugs in the field of T2D. Such drugs would be of great interest since they can be used both in the treatment of diabetes and obesity. This patent review aims to establish an overview of recent patents disclosing new therapeutic opportunities targeting peripheral, as well as central targets for the treatment of T2D.
Collapse
|
12
|
Ding H, Zhu T, Yin X, Liu J, Zhang L, Bernier M, Zhao R. Pyrrolidine dithiocarbamate protects pancreatic β-cells from oxidative damage through regulation of FoxO1 activity in type 2 diabetes rats. Acta Biochim Biophys Sin (Shanghai) 2014; 46:582-9. [PMID: 24829401 DOI: 10.1093/abbs/gmu034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pyrrolidine dithiocarbamate (PDTC) can lower the blood glucose level and improve the insulin sensitivity in diabetic rats. However, the mechanisms underlying this effect of PDTC treatment in diabetic rats remained uncertain. In this study, we evaluated the mechanisms by which PDTC conferred protection against oxidative damage to pancreatic islet β-cells in rats with experimental type 2 diabetes mellitus (DM). DM in the rats was elicited by long-term high-fat diet accompanied with a single intraperitoneal (i.p.) injection of a low dose of streptozotocin. After a 7-day administration of PDTC (50 mg/kg/day i.p.), blood glucose levels were measured and pancreatic tissues were collected for the determination of various biochemical and enzymatic activities using immunohistochemistry, immunofluorescence, and western blot techniques. The percentage of apoptotic pancreatic islet β-cells was detected by flow cytometry. The results showed that diabetic rats had elevated blood glucose levels and insulin resistance, accompanied with an increase in malondialdehyde content, nitrotyrosine production, and inducible nitric oxide synthase expression. A decrease in superoxide dismutase and glutathione peroxidase activities was also observed in DM rats, culminating with elevated β-cell apoptosis. PDTC treatment significantly reduced the oxidative damage and the β-cell apoptosis, and also increased the insulin production through down-regulating FoxO1 acetylation and up-regulating nuclear PDX-1 level. These data suggested that PDTC can protect islet β-cells from oxidative damage and improve insulin production through regulation of PDX-1 and FoxO1 in a DM rat model.
Collapse
Affiliation(s)
- Haiyan Ding
- Department of Endocrinology, Fourth Hospital, Hebei Medical University, Shijiazhuang 050011, China
| | - Tienian Zhu
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang 050017, China Department of Medical Oncology, Bethune International Peace Hospital, Shijiazhuang 050082, China
| | - Xiaomei Yin
- Department of Medical Oncology, Bethune International Peace Hospital, Shijiazhuang 050082, China
| | - Jiankun Liu
- Department of Medical Oncology, Bethune International Peace Hospital, Shijiazhuang 050082, China
| | - Lizhong Zhang
- Department of Medical Oncology, Bethune International Peace Hospital, Shijiazhuang 050082, China
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ruijing Zhao
- Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Department of Immunology, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
13
|
Merry TL, Tran M, Stathopoulos M, Wiede F, Fam BC, Dodd GT, Clarke I, Watt MJ, Andrikopoulos S, Tiganis T. High-fat-fed obese glutathione peroxidase 1-deficient mice exhibit defective insulin secretion but protection from hepatic steatosis and liver damage. Antioxid Redox Signal 2014; 20:2114-29. [PMID: 24252128 DOI: 10.1089/ars.2013.5428] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AIMS Reactive oxygen species (ROS) such as H2O2 can promote signaling through the inactivation of protein tyrosine phosphatases (PTPs). However, in obesity, the generation of ROS exceeds the antioxidant reserve and can contribute to the promotion of insulin resistance. Glutathione peroxidase 1 (Gpx1) is an antioxidant enzyme that eliminates H2O2. Here, we have used Gpx1(-/-) mice to assess the impact of oxidative stress on glucose homeostasis in the context of obesity. RESULTS Gpx1(-/-) mice fed an obesogenic high-fat diet for 12 weeks exhibited systemic oxidative stress and hyperglycemia, but had unaltered whole-body insulin sensitivity, improved hepatic insulin signaling, and decreased whole-body glucose production. High-fat-fed Gpx1(-/-) mice also exhibited decreased hepatic steatosis and liver damage accompanied by decreased plasma insulin and decreased glucose-induced insulin secretion. The decreased insulin secretion was associated with reduced islet β cell pancreatic and duodenal homeobox-1 (Pdx1) and insulin content, elevated pancreatic PTP oxidation (including PTPN2 oxidation), and elevated signal transducer and activator of transcription 1 (STAT1) Y701 phosphorylation. INNOVATION AND CONCLUSION Taken together, these results are consistent with H2O2 inactivating pancreatic PTPs (such as the STAT1 phosphatase PTPN2) for the promotion of STAT-1 signaling to suppress Pdx1 expression and differentiation and, consequently, reduce β cell insulin secretion. We propose that the decreased insulin secretion, in turn, results in decreased hepatic lipogenesis and steatosis, attenuates liver damage, and improves hepatic insulin signaling to suppress hepatic glucose production. Limiting insulin secretion may help combat the development of hepatic steatosis and liver damage in diet-induced obesity.
Collapse
Affiliation(s)
- Troy L Merry
- 1 Department of Biochemistry and Molecular Biology, Monash University , Victoria, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Tveden-Nyborg P, Lykkesfeldt J. Does vitamin C deficiency increase lifestyle-associated vascular disease progression? Evidence based on experimental and clinical studies. Antioxid Redox Signal 2013; 19:2084-104. [PMID: 23642093 DOI: 10.1089/ars.2013.5382] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Despite continuous advances in the prevention of cardiovascular disease (CVD), critical issues associated with an unhealthy lifestyle remain an increasing cause of morbidity and mortality in industrialized countries. RECENT ADVANCES A growing body of literature supports a specific role for vitamin C in a number of reactions that are associated with vascular function and control including, for example, nitric oxide bioavailability, lipid metabolism, and vascular integrity. CRITICAL ISSUES A large body of epidemiological evidence supports a relationship between poor vitamin C status and increased risk of developing CVD, and the prevalence of deficiency continues to be around 10%-20% of the general Western population although this problem could easily and cheaply be solved by supplementation. However, large intervention studies using vitamin C have not found a beneficial effect of supplementation. This review outlines the proposed mechanism by which vitamin C deficiency worsens CVD progression. In addition, it discusses problems with the currently available literature, including the discrepancies between the large intervention studies and the experimental and epidemiological literature. FUTURE DIRECTIONS Increased insights into vitamin C deficiency-mediated CVD progression will enable the design of future randomized controlled trials that are better suited to test the efficacy of vitamin C in disease prevention as well as the identification of high-risk individuals which could possibly benefit from supplementation.
Collapse
Affiliation(s)
- Pernille Tveden-Nyborg
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen , Frederiksberg, Denmark
| | | |
Collapse
|
15
|
Wong N, Morahan G, Stathopoulos M, Proietto J, Andrikopoulos S. A novel mechanism regulating insulin secretion involving Herpud1 in mice. Diabetologia 2013; 56:1569-76. [PMID: 23620059 DOI: 10.1007/s00125-013-2908-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 03/18/2013] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes results from beta cell dysfunction after prolonged physiological stress, which causes oversecretion of insulin. We recently found that insulin hypersecretion is mediated by at least two genes. Among mouse models of type 2 diabetes, the DBA/2 mouse strain is more susceptible to diabetes than is the C57BL/6J (B6J) strain. One distinctive feature of the DBA/2 mouse is that it hypersecretes insulin, independent of changes in insulin sensitivity; we identified Nnt as a gene responsible for this trait. METHODS To identify the other gene(s) affecting insulin hypersecretion, we tested a panel of recombinant inbred BXD strains, which have different combinations of B6 and DBA/2 alleles. RESULTS We found that 25% of the BXD strains hypersecreted insulin in response to glucose. Microarray profiling of islets from high- and low-secretor strains showed that at least four genes were differentially expressed. One gene was consistently underexpressed in islets from both DBA/2 and the high-secretor BXD strains. This gene (Herpud1 or Herp) encodes the 54 kDa endoplasmic reticulum stress-inducible protein (HERP) that resides in the integral endoplasmic reticulum membrane. To test directly whether Herpud1 can interact with Nnt, Herpud1 was either knocked down or overexpressed in MIN6 cells. These results showed that when Herpud1 was suppressed, Nnt expression was reduced, while overexpression of Herpud1 led to increased Nnt expression. Furthermore, Herpud1 suppression resulted in significantly decreased glucose-stimulated insulin secretion in the DBA/2 islets but not B6J islets. CONCLUSIONS/INTERPRETATION We conclude that Herpud1 regulates insulin secretion via control of Nnt expression.
Collapse
Affiliation(s)
- N Wong
- Department of Medicine (Austin Health), Austin Hospital, University of Melbourne, Heidelberg Heights, Melbourne, Victoria, 3084, Australia.
| | | | | | | | | |
Collapse
|
16
|
Efficacy of azelaic acid on hepatic key enzymes of carbohydrate metabolism in high fat diet induced type 2 diabetic mice. Biochimie 2013; 95:1239-44. [DOI: 10.1016/j.biochi.2013.01.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 01/29/2013] [Indexed: 11/23/2022]
|
17
|
Lamontagne J, Jalbert-Arsenault É, Pepin É, Peyot ML, Ruderman NB, Nolan CJ, Joly E, Madiraju SM, Poitout V, Prentki M. Pioglitazone acutely reduces energy metabolism and insulin secretion in rats. Diabetes 2013; 62:2122-9. [PMID: 23378607 PMCID: PMC3661607 DOI: 10.2337/db12-0428] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our objective was to determine if the insulin-sensitizing drug pioglitazone acutely reduces insulin secretion and causes metabolic deceleration in vivo independently of change in insulin sensitivity. We assessed glucose homeostasis by hyperinsulinemic-euglycemic and hyperglycemic clamp studies and energy expenditure by indirect calorimetry and biotelemetry in male Wistar and obese hyperinsulinemic Zucker diabetic fatty (ZDF) rats 45 min after a single oral dose of pioglitazone (30 mg/kg). In vivo insulin secretion during clamped hyperglycemia was reduced in both Wistar and ZDF rats after pioglitazone administration. Insulin clearance was slightly increased in Wistar but not in ZDF rats. Insulin sensitivity in Wistar rats assessed by the hyperinsulinemic-euglycemic clamp was minimally affected by pioglitazone at this early time point. Pioglitazone also reduced energy expenditure in Wistar rats without altering respiratory exchange ratio or core body temperature. Glucose-induced insulin secretion (GIIS) and oxygen consumption were reduced by pioglitazone in isolated islets and INS832/13 cells. In conclusion, pioglitazone acutely induces whole-body metabolic slowing down and reduces GIIS, the latter being largely independent of the insulin-sensitizing action of the drug. The results suggest that pioglitazone has direct metabolic deceleration effects on the β-cell that may contribute to its capacity to lower insulinemia and antidiabetic action.
Collapse
Affiliation(s)
- Julien Lamontagne
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
| | - Élise Jalbert-Arsenault
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Émilie Pepin
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Program of Molecular Biology, Université de Montréal, Montreal, Quebec, Canada
| | - Marie-Line Peyot
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Neil B. Ruderman
- Departments of Medicine and Physiology and Biophysics, Boston University School of Medicine and Diabetes Unit, Section of Endocrinology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts
| | | | - Erik Joly
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - S.R. Murthy Madiraju
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
| | - Vincent Poitout
- Montreal Diabetes Research Center at the CRCHUM and Departments of Medicine, Biochemistry and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Marc Prentki
- Molecular Nutrition Unit and Montreal Diabetes Research Center at the Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Université de Montréal, Montreal, Quebec, Canada
- Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, Université de Montréal, Montreal, Quebec, Canada
- Corresponding author: Marc Prentki,
| |
Collapse
|
18
|
Loizzo A, Spampinato SM, Campana G, Vella S, Fortuna A, Costa L, Capasso A, Monteleone P, Renzi P, Loizzo S. Enhanced brain performance in mice following postnatal stress. J Endocrinol 2012; 215:413-24. [PMID: 23045190 DOI: 10.1530/joe-12-0369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The double postnatal stress model (brief maternal separation plus sham injection daily applied from birth to weaning) induces metabolic alterations similar to type 2 diabetes in young-adult male mice. We verify whether 1) the stress also induces brain metabolic-functional alterations connected to diabetes and 2) different alterations are modulated selectively by two stress-damaged endogenous systems (opioid- and/or ACTH-corticosteroid-linked). Here, diabetes-like metabolic plus neurophysiologic-neurometabolic parameters are studied in adult mice following postnatal stress and drug treatment. Surprisingly, together with 'classic' diabetes-like alterations, the stress model induces in young-adult mice significantly enhanced brain neurometabolic-neurophysiologic performances, consisting of decreased latency to flash-visual evoked potentials (- ~8%); increased level (+ ~40%) and reduced latency (- ~30%) of NAD(P)H autofluorescence postsynaptic signals following electric stimuli; enhanced passive avoidance learning (+ ~135% latency); and enhanced brain-derived neurotrophic factor level (+ ~70%). Postnatal treatment with the opioid receptor antagonist naloxone prevents some alterations, moreover the treatment with antisense (AS; AS vs proopiomelanocortin mRNA) draws all parameters to control levels, thus showing that some alterations are bound to endogenous opioid-system hyper-functioning, while others depend on ACTH-corticosterone system hyper-functioning. Our stress model induces diabetes-like metabolic alterations coupled to enhanced brain neurometabolic-neurophysiologic performances. Taken all together, these findings are compatible with an 'enduring acute-stress' reaction, which puts mice in favorable survival situations vs controls. However, prolonged hormonal-metabolic imbalances are expected to also produce diabetes-like complications at later ages in stressed mice.
Collapse
Affiliation(s)
- Alberto Loizzo
- Department of Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanita', via Regina Elena 299, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Avignon A, Hokayem M, Bisbal C, Lambert K. Dietary antioxidants: Do they have a role to play in the ongoing fight against abnormal glucose metabolism? Nutrition 2012; 28:715-21. [DOI: 10.1016/j.nut.2012.01.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 01/04/2012] [Accepted: 01/04/2012] [Indexed: 01/01/2023]
|
20
|
Webster BR, Lu Z, Sack MN, Scott I. The role of sirtuins in modulating redox stressors. Free Radic Biol Med 2012; 52:281-90. [PMID: 22085655 PMCID: PMC3253188 DOI: 10.1016/j.freeradbiomed.2011.10.484] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/24/2011] [Accepted: 10/25/2011] [Indexed: 12/21/2022]
Abstract
For much of the time since their discovery, the sirtuin family of deacetylase enzymes has been associated with extension of life span. This longevity-promoting capacity in numerous model systems has enabled the sirtuins to gain "celebrity status" in the field of aging research. However, the mechanisms underpinning these changes remain incompletely defined. A general phenotype long associated with aging is the dysregulation of biological systems, which partly occurs via the accumulation of damage over time. One of the major sources of this damage is oxidative stress, which can harm both biological structures and the mechanisms with which they are repaired. It is now becoming clear that the beneficial life-span effects of sirtuins, along with many of their other functions, are closely linked to their ability to regulate systems that control the redox environment. Here we investigate the links between sirtuins and their oxidative/redox environment and review the control mechanisms that are regulated by the activity of sirtuin deacetylase proteins.
Collapse
Affiliation(s)
- Bradley R Webster
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
21
|
Iwasaki S, Widjaja-Adhi MAK, Koide A, Kaga T, Nakano S, Beppu F, Hosokawa M, Miyashita K. <i>In Vivo</i> Antioxidant Activity of Fucoxanthin on Obese/Diabetes KK-<i>A<sup>y</sup></i> Mice. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/fns.2012.311194] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Ishizuka M, Kubota K, Kita J, Shimoda M, Kato M, Sawada T. Underweight patients show an increased rate of postoperative death after surgery for hepatocellular carcinoma. J Surg Oncol 2011; 104:809-13. [PMID: 21815148 DOI: 10.1002/jso.22049] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Accepted: 07/05/2011] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate the influence of body mass index (BMI) on postoperative death in patients undergoing surgery for HCC. METHODS Three hundred forty-two patients were enrolled, and divided into three groups: Group A, BMI <22.5; Group B, BMI ≥22.5 to <25; Group C, BMI ≥25. Univariate and multivariate analyses of postoperative death were performed to compare BMI with clinical factors. Kaplan-Meier analysis and log rank test were used to compare such outcome in Groups A, B, and C. RESULTS Kaplan-Meier analysis and log rank test revealed that Group A had a higher rate of postoperative death than Group B or C (P = 0.010). Univariate and multivariate analyses selected being underweight (Group B, C/Group A) (odds ratio, 1.829; 95% C.I., 1.091-3.068; P = 0.022) as one of the factors predictive of postoperative death, together with aspartate aminotransferase level (P = 0.042) and HCC growth pattern (P = 0.032). CONCLUSIONS BMI is a simple but important predictor of postoperative death in patients undergoing surgery for HCC, and is able to classify such patients into three independent groups.
Collapse
Affiliation(s)
- Mitsuru Ishizuka
- Department of Gastroenterological Surgery, Dokkyo Medical University, Mibu, Tochigi, Japan.
| | | | | | | | | | | |
Collapse
|
23
|
Bibliography. Current world literature. Adrenal cortex. Curr Opin Endocrinol Diabetes Obes 2011; 18:231-3. [PMID: 21522003 DOI: 10.1097/med.0b013e3283457c7d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Rajpal DK, Kumar V, Agarwal P. Scientific literature mining for drug discovery: a case study on obesity. Drug Dev Res 2010. [DOI: 10.1002/ddr.20416] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|