1
|
Das M, Banerjee A, Roy R. A novel in vitro approach to test the effectiveness of fish oil in ameliorating type 1 diabetes. Mol Cell Biochem 2022; 477:2121-2132. [PMID: 35545742 DOI: 10.1007/s11010-022-04424-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 03/25/2022] [Indexed: 11/29/2022]
Abstract
Diabetes type 1 (T1D) characterized by destruction of pancreatic β-cells results in inadequate insulin production and hyperglycaemia. Generation of reactive oxygen species and glycosylation end-products stimulates toxic impacts on T1D. Dietary w-3 fatty acids present in Fish oil (FO) might be helpful in the prevention of oxidative stress and lipid peroxidation, thus, beneficial against T1D. But how the cellular secretion from β-cells under influence of FO affects the glucose homeostasis of peri-pancreatic cells is poorly understood. In the current study, we aimed to introduce an in vitro model for T1D and evaluate its effectiveness in respect of alloxan treatment to pancreatic Min6 cells. We use alloxan in the Min6 pancreatic β-cell line to induce cellular damage related to T1D. Further treatment with FO was seen to prevent cell death by alloxan and induce mRNA expression of both insulin 1 and insulin 2 isoforms under low-glucose conditions. From the first part of the study, it is clear that FO is effective to recover Min6 cells from the destructive effect of alloxan, and it worked best when given along with alloxan or given after alloxan treatment regime. FO-induced secretion of molecules from Min6 was clearly shown to regulate mRNA expression of key enzymes of carbohydrate metabolism in peri-pancreatic cell types. This is a pilot study showing that an improved in vitro approach of using Min6 along with muscle cells (C2C12) and adipose tissue cells (3T3-L1) together to understand the crosstalk of molecules could be used to check the efficacy of an anti-diabetic drug.
Collapse
Affiliation(s)
- Moitreyi Das
- Department of Zoology, Goa University, Goa, India
| | - Arnab Banerjee
- Department of Biological Sciences, BITS Pilani, K. K. Birla Goa Campus, Zuarinagar, Goa, India
| | | |
Collapse
|
2
|
Dietary Protein Modulates the Efficacy of Taurine Supplementation on Adaptive Islet Function and Morphology in Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1370:267-278. [DOI: 10.1007/978-3-030-93337-1_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
3
|
Freitas-Dias R, Lima TI, Costa-Junior JM, Gonçalves LM, Araujo HN, Paula FMM, Santos GJ, Branco RCS, Ou K, Kaestner KH, Silveira LR, Oliveira CAM, Boschero AC, Zoppi CC, Carneiro EM. Offspring from trained male mice inherit improved muscle mitochondrial function through PPAR co-repressor modulation. Life Sci 2021; 291:120239. [PMID: 34942163 DOI: 10.1016/j.lfs.2021.120239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 10/19/2022]
Abstract
Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.
Collapse
Affiliation(s)
- Ricardo Freitas-Dias
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Physical Therapy, Laboratory of Exercise Physiology, University of Pernambuco, Petrolina, PE, Brazil
| | - Tanes I Lima
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jose Maria Costa-Junior
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luciana M Gonçalves
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Flavia M M Paula
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Gustavo J Santos
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianopolis, SC, Brazil
| | - Renato Chaves Souto Branco
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Kristy Ou
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Camila A M Oliveira
- Department of Biosciences, Federal University of Sao Paulo, Santos, SP, Brazil
| | - Antonio C Boschero
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Claudio C Zoppi
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Everardo M Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
4
|
Damame HH, Rooge SB, Patil RS, Arvindekar AU. In vitro model using cytokine cocktail to evaluate apoptosis in Min6 pancreatic beta cells. J Pharmacol Toxicol Methods 2020; 106:106914. [PMID: 32828949 DOI: 10.1016/j.vascn.2020.106914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/08/2020] [Accepted: 08/13/2020] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Development of therapy options for treatment of type 1 diabetes mellitus is hampered by non-availability of appropriate experimental models that can exactly mimic the in vivo situation. Apoptosis of beta cells by T cells and cytokine action leads to loss of beta cells. We propose a simple and elegant model using cytokine cocktail of TNF-α, IFN-γ and IL-1β, the major cytokines responsible for apoptosis in Min6 beta cell line. METHODS A cocktail of TNF-α, IFN-γ and IL-1β was used to induce apoptosis in Min6 beta cell line. Apoptosis was assessed by flow cytometry using CytoFLEX (Beckman Coulter). The destruction of beta cells is through production of nitric oxide (NO), oxidative stress and change in mitochondrial membrane permeability. NO was measured using Griess reagent. Oxidative stress was assessed using 2',7'-dichlorofluorescein diacetate, a cell-permeable fluorogenic dye and mitochondrial membrane potential was determined on the basis of retention of rhodamine 123 using flow cytometer. RESULTS AND DISCUSSION Very low concentration of the cocktail viz. TNF-α 25 ng/ml, IFN-γ 25 ng/ml and IL-1β 50 ng/ml has demonstrated effective early and late apoptosis in as short a time period as 6 h. The experimental model used demonstrated 1.5 fold higher production of NO, 1.2 fold increased oxidative stress and lower mitochondrial membrane potential as compared to the positive control used. Hence the above model can be easily used for assessment and screening of drugs that can prevent apoptosis of beta cells and stop progression of type 1 diabetes.
Collapse
Affiliation(s)
- Hemangee H Damame
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India
| | - Sheetalnath B Rooge
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India
| | - Rahul S Patil
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India
| | - Akalpita U Arvindekar
- Department of Biochemistry, Shivaji University, Kolhapur 416 004, Maharashtra, India.
| |
Collapse
|
5
|
Qi HY, Li L, Ma H. Cellular stress response mechanisms as therapeutic targets of ginsenosides. Med Res Rev 2017; 38:625-654. [DOI: 10.1002/med.21450] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/28/2017] [Accepted: 04/14/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Hong-yi Qi
- College of Chinese Medicine; Southwest University; Chongqing P.R. China
| | - Li Li
- College of Chinese Medicine; Southwest University; Chongqing P.R. China
| | - Hui Ma
- College of Chinese Medicine; Southwest University; Chongqing P.R. China
| |
Collapse
|
6
|
Branco RCS, Camargo RL, Batista TM, Vettorazzi JF, Borck PC, Dos Santos-Silva JCR, Boschero AC, Zoppi CC, Carneiro EM. Protein malnutrition blunts the increment of taurine transporter expression by a high-fat diet and impairs taurine reestablishment of insulin secretion. FASEB J 2017; 31:4078-4087. [PMID: 28572444 DOI: 10.1096/fj.201600326rrr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 05/15/2017] [Indexed: 12/23/2022]
Abstract
Taurine (Tau) restores β-cell function in obesity; however, its action is lost in malnourished obese rodents. Here, we investigated the mechanisms involved in the lack of effects of Tau in this model. C57BL/6 mice were fed a control diet (CD) (14% protein) or a protein-restricted diet (RD) (6% protein) for 6 wk. Afterward, mice received a high-fat diet (HFD) for 8 wk [CD + HFD (CH) and RD + HFD (RH)] with or without 5% Tau supplementation after weaning on their drinking water [CH + Tau (CHT) and RH + Tau (RHT)]. The HFD increased insulin secretion through mitochondrial metabolism in CH and RH. Tau prevented all those alterations in CHT only. The expression of the taurine transporter (Tau-T), as well as Tau content in pancreatic islets, was increased in CH but had no effect on RH. Protein malnutrition programs β cells and impairs Tau-induced restoration of mitochondrial metabolism and biogenesis. This may be associated with modulation of the expression of Tau-T in pancreatic islets, which may be responsible for the absence of effect of Tau in protein-malnourished obese mice.-Branco, R. C. S., Camargo, R. L., Batista, T. M., Vettorazzi, J. F., Borck, P. C., dos Santos-Silva, J. C. R., Boschero, A. C., Zoppi, C. C., Carneiro, E. M. Protein malnutrition blunts the increment of taurine transporter expression by a high-fat diet and impairs taurine reestablishment of insulin secretion.
Collapse
Affiliation(s)
- Renato Chaves Souto Branco
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Rafael Ludemann Camargo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Thiago Martins Batista
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Jean Franciesco Vettorazzi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Patrícia Cristine Borck
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | - Antonio Carlos Boschero
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Cláudio Cesar Zoppi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Everardo Magalhães Carneiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
7
|
Acute Exercise Improves Insulin Clearance and Increases the Expression of Insulin-Degrading Enzyme in the Liver and Skeletal Muscle of Swiss Mice. PLoS One 2016; 11:e0160239. [PMID: 27467214 PMCID: PMC4965115 DOI: 10.1371/journal.pone.0160239] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 07/15/2016] [Indexed: 11/19/2022] Open
Abstract
The effects of exercise on insulin clearance and IDE expression are not yet fully elucidated. Here, we have explored the effect of acute exercise on insulin clearance and IDE expression in lean mice. Male Swiss mice were subjected to a single bout of exercise on a speed/angle controlled treadmill for 3-h at approximately 60-70% of maximum oxygen consumption. As expected, acute exercise reduced glycemia and insulinemia, and increased insulin tolerance. The activity of AMPK-ACC, but not of IR-Akt, pathway was increased in the liver and skeletal muscle of trained mice. In an apparent contrast to the reduced insulinemia, glucose-stimulated insulin secretion was increased in isolated islets of these mice. However, insulin clearance was increased after acute exercise and was accompanied by increased expression of the insulin-degrading enzyme (IDE), in the liver and skeletal muscle. Finally, C2C12, but not HEPG2 cells, incubated at different concentrations of 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) for 3-h, showed increased expression of IDE. In conclusion, acute exercise increases insulin clearance, probably due to an augmentation of IDE expression in the liver and skeletal muscle. The elevated IDE expression, in the skeletal muscle, seems to be mediated by activation of AMPK-ACC pathway, in response to exercise. We believe that the increase in the IDE expression, comprise a safety measure to maintain glycemia at or close to physiological levels, turning physical exercise more effective and safe.
Collapse
|
8
|
Fred RG, Kappe C, Ameur A, Cen J, Bergsten P, Ravassard P, Scharfmann R, Welsh N. Role of the AMP kinase in cytokine-induced human EndoC-βH1 cell death. Mol Cell Endocrinol 2015. [PMID: 26213325 DOI: 10.1016/j.mce.2015.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The aim of the present investigation was to delineate cytokine-induced signaling and death using the EndoC-βH1 cells as a model for primary human beta-cells. The cytokines IL-1β and IFN-γ induced a rapid and transient activation of NF-κB, STAT-1, ERK, JNK and eIF-2α signaling. The EndoC-βH1 cells died rapidly when exposed to IL-1β + IFN-γ, and this occurred also in the presence of the actinomycin D. Inhibition of NF-κB and STAT-1 did not protect against cell death, nor did the cytokines activate iNOS expression. Instead, cytokines promoted a rapid decrease in EndoC-βH1 cell respiration and ATP levels, and we observed protection by the AMPK activator AICAR against cytokine-induced cell death. It is concluded that EndoC-βH1 cell death can be prevented by AMPK activation, which suggests a role for ATP depletion in cytokine-induced human beta-cell death.
Collapse
Affiliation(s)
- Rikard G Fred
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Camilla Kappe
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Adam Ameur
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, SE-75185 Uppsala, Sweden
| | - Jing Cen
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Peter Bergsten
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden
| | - Phillippe Ravassard
- Biotechnology and Biotherapy Laboratory, Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, CHU Pitié-Salpêtrière, Paris, France
| | - Raphael Scharfmann
- INSERM, U1016, Institut Cochin, Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Nils Welsh
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Box 571, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
9
|
Tong X, Kono T, Evans-Molina C. Nitric oxide stress and activation of AMP-activated protein kinase impair β-cell sarcoendoplasmic reticulum calcium ATPase 2b activity and protein stability. Cell Death Dis 2015; 6:e1790. [PMID: 26086963 PMCID: PMC4669835 DOI: 10.1038/cddis.2015.154] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 04/13/2015] [Accepted: 04/20/2015] [Indexed: 11/15/2022]
Abstract
The sarcoendoplasmic reticulum Ca2+ ATPase 2b (SERCA2b) pump maintains a steep Ca2+ concentration gradient between the cytosol and ER lumen in the pancreatic β-cell, and the integrity of this gradient has a central role in regulated insulin production and secretion, maintenance of ER function and β-cell survival. We have previously demonstrated loss of β-cell SERCA2b expression under diabetic conditions. To define the mechanisms underlying this, INS-1 cells and rat islets were treated with the proinflammatory cytokine interleukin-1β (IL-1β) combined with or without cycloheximide or actinomycin D. IL-1β treatment led to increased inducible nitric oxide synthase (iNOS) gene and protein expression, which occurred concurrently with the activation of AMP-activated protein kinase (AMPK). IL-1β led to decreased SERCA2b mRNA and protein expression, whereas time-course experiments revealed a reduction in protein half-life with no change in mRNA stability. Moreover, SERCA2b protein but not mRNA levels were rescued by treatment with the NOS inhibitor l-NMMA (NG-monomethyl l-arginine), whereas the NO donor SNAP (S-nitroso-N-acetyl-d,l-penicillamine) and the AMPK activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) recapitulated the effects of IL-1β on SERCA2b protein stability. Similarly, IL-1β-induced reductions in SERCA2b expression were rescued by pharmacological inhibition of AMPK with compound C or by transduction of a dominant-negative form of AMPK, whereas β-cell death was prevented in parallel. Finally, to determine a functional relationship between NO and AMPK signaling and SERCA2b activity, fura-2/AM (fura-2-acetoxymethylester) Ca2+ imaging experiments were performed in INS-1 cells. Consistent with observed changes in SERCA2b expression, IL-1β, SNAP and AICAR increased cytosolic Ca2+ and decreased ER Ca2+ levels, suggesting congruent modulation of SERCA activity under these conditions. In aggregate, these results show that SERCA2b protein stability is decreased under inflammatory conditions through NO- and AMPK-dependent pathways and provide novel insight into pathways leading to altered β-cell calcium homeostasis and reduced β-cell survival in diabetes.
Collapse
Affiliation(s)
- X Tong
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - T Kono
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - C Evans-Molina
- 1] Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA [2] Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA [3] Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA [4] The Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA [5] The Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
10
|
Costa-Júnior JM, Ferreira SM, Protzek AO, Santos GJ, Cappelli AP, Silveira LR, Zoppi C, de Oliveira CAM, Boschero AC, Carneiro EM, Rezende LF. Endurance training inhibits insulin clearance and IDE expression in Swiss mice. PLoS One 2015; 10:e0118809. [PMID: 25822220 PMCID: PMC4379169 DOI: 10.1371/journal.pone.0118809] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 01/08/2015] [Indexed: 01/26/2023] Open
Abstract
INTRODUCTION Endurance training improves peripheral insulin sensitivity in the liver and the skeletal muscle, but the mechanism for this effect is poorly understood. Recently, it was proposed that insulin clearance plays a major role in both glucose homeostasis and insulin sensitivity. Therefore, our goal was to determine the mechanism by which endurance training improves insulin sensitivity and how it regulates insulin clearance in mice. METHODS Mice were treadmill-trained for 4 weeks at 70-80% of maximal oxygen consumption (VO2 max) for 60 min, 5 days a week. The glucose tolerance and the insulin resistance were determined using an IPGTT and an IPITT, respectively, and the insulin decay rate was calculated from the insulin clearance. Protein expression and phosphorylation in the liver and the skeletal muscle were ascertained by Western blot. RESULTS Trained mice exhibited an increased VO2 max, time to exhaustion, glucose tolerance and insulin sensitivity. They had smaller fat pads and lower plasma concentrations of insulin and glucose. Endurance training inhibited insulin clearance and reduced expression of IDE in the liver, while also inhibiting insulin secretion by pancreatic islets. There was increased phosphorylation of both the canonical (IR-AKT) and the non-canonical (CaMKII-AMPK-ACC) insulin pathways in the liver of trained mice, whereas only the CaMKII-AMPK pathway was increased in the skeletal muscle. CONCLUSION Endurance training improved glucose homeostasis not only by increasing peripheral insulin sensitivity but also by decreasing insulin clearance and reducing IDE expression in the liver.
Collapse
Affiliation(s)
- José M. Costa-Júnior
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Sandra M. Ferreira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - André O. Protzek
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Gustavo J. Santos
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Ana P. Cappelli
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Leonardo R. Silveira
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of Sao Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Cláudio Zoppi
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Camila A. M. de Oliveira
- Department of Biosciences, Federal University of Sao Paulo (Unifesp), Santos, SP, CEP 11060-001, Brazil
| | - Antonio C. Boschero
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Everardo M. Carneiro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | - Luiz F. Rezende
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, SP, CEP 13083-865, Brazil
- * E-mail:
| |
Collapse
|
11
|
Ferreira SM, Santos GJ, Rezende LF, Gonçalves LM, Santos-Silva JC, Bigarella CL, Carneiro EM, Saad STO, Boschero AC, Barbosa-Sampaio HC. ARHGAP21 prevents abnormal insulin release through actin rearrangement in pancreatic islets from neonatal mice. Life Sci 2015; 127:53-8. [PMID: 25744409 DOI: 10.1016/j.lfs.2015.01.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/06/2015] [Accepted: 01/26/2015] [Indexed: 12/31/2022]
Abstract
AIMS ARHGAP21 is a Rho GTPase-activating protein (RhoGAP) that associates with many proteins and modulates several cellular functions, including actin cytoskeleton rearrangement in different tissues. However, it is unknown whether ARHGAP21 is expressed in pancreatic beta cells and its function in these cells. Herein, we assess the participation of ARHGAP21 in insulin secretion. MAIN METHODS Neonatal mice were treated with anti-sense oligonucleotide against ARHG AP21 (AS) for 2 days, resulting in a reduction of the protein's expression of about 60% in the islets. F-actin depolimerization, insulin secretion,mRNA level of genes involved in insulin secretion, maturation and proliferation were evaluated in islets from both control and AS-treated mice. KEY FINDINGS ARHGAP21 co-localized with actin inMIN6 beta cells and with insulin in neonatal pancreatic islets. F-actin was reduced in AS-islets, as judged by lower phalloidin intensity. Insulin secretion was increased in islets from AS-treated mice, however no differences were observed in the GSIS (glucose-stimulated insulin secretion). In these islets, the pERK1/2 was increased, as well as the gene expressions of VAMP2 and SNAP25, proteins that are present in the secretory machinery. Maturation and cell proliferation were not affected in islets from AS-treated mice. SIGNIFICANCE In conclusion, our data show, for the first time, that ARHGAP21 is expressed and participates in the secretory process of pancreatic beta cells. Its effect is probably via pERK1/2, which modulates the rearrangement of the cytoskeleton. ARHGAP21 also controls the expression of genes that encodes proteins of the secretory machinery.
Collapse
Affiliation(s)
- Sandra Mara Ferreira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Gustavo Jorge Santos
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luiz F Rezende
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luciana Mateus Gonçalves
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Junia Carolina Santos-Silva
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Carolina Louzão Bigarella
- Department of Internal Medicine, School of Medical Science, Hematology and Hemotherapy Center - Hemocentro, INCT Sangue, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Everardo Magalhães Carneiro
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Sara Teresinha Ollala Saad
- Department of Internal Medicine, School of Medical Science, Hematology and Hemotherapy Center - Hemocentro, INCT Sangue, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Antonio Carlos Boschero
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Helena Cristina Barbosa-Sampaio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
12
|
Camargo RL, Branco RCS, de Rezende LF, Vettorazzi JF, Borck PC, Boschero AC, Carneiro EM. The Effect of Taurine Supplementation on Glucose Homeostasis: The Role of Insulin-Degrading Enzyme. TAURINE 9 2015; 803:715-24. [DOI: 10.1007/978-3-319-15126-7_57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
13
|
Reduced insulin clearance and lower insulin-degrading enzyme expression in the liver might contribute to the thrifty phenotype of protein-restricted mice. Br J Nutr 2014; 112:900-7. [PMID: 25036874 DOI: 10.1017/s0007114514001238] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Nutrient restriction during the early stages of life usually leads to alterations in glucose homeostasis, mainly insulin secretion and sensitivity, increasing the risk of metabolic disorders in adulthood. Despite growing evidence regarding the importance of insulin clearance during glucose homeostasis in health and disease, no information exists about this process in malnourished animals. Thus, in the present study, we aimed to determine the effect of a nutrient-restricted diet on insulin clearance using a model in which 30-d-old C57BL/6 mice were exposed to a protein-restricted diet for 14 weeks. After this period, we evaluated many metabolic variables and extracted pancreatic islet, liver, gastrocnemius muscle (GCK) and white adipose tissue samples from the control (normal-protein diet) and restricted (low-protein diet, LP) mice. Insulin concentrations were determined using RIA and protein expression and phosphorylation by Western blot analysis. The LP mice exhibited lower body weight, glycaemia, and insulinaemia, increased glucose tolerance and altered insulin dynamics after the glucose challenge. The improved glucose tolerance could partially be explained by an increase in insulin sensitivity through the phosphorylation of the insulin receptor/protein kinase B and AMP-activated protein kinase/acetyl-CoA carboxylase in the liver, whereas the changes in insulin dynamics could be attributed to reduced insulin secretion coupled with reduced insulin clearance and lower insulin-degrading enzyme (IDE) expression in the liver and GCK. In summary, protein-restricted mice not only produce and secrete less insulin, but also remove and degrade less insulin. This phenomenon has the double benefit of sparing insulin while prolonging and potentiating its effects, probably due to the lower expression of IDE in the liver, possibly with long-term consequences.
Collapse
|
14
|
Guan FY, Gu J, Li W, Zhang M, Ji Y, Li J, Chen L, Hatch GM. Compound K protects pancreatic islet cells against apoptosis through inhibition of the AMPK/JNK pathway in type 2 diabetic mice and in MIN6 β-cells. Life Sci 2014; 107:42-9. [PMID: 24802125 DOI: 10.1016/j.lfs.2014.04.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 04/08/2014] [Accepted: 04/24/2014] [Indexed: 01/27/2023]
Abstract
AIMS Compound K (CK) is known to possess anti-diabetic activities but the mechanism for this action is unknown. The present study observed the protective effect of CK on islet cell apoptosis through the AMP-activated protein kinase (AMPK) mediated C-Jun N-terminal kinase (JNK) pathway. MAIN METHODS Treatment effect of CK on type 2 diabetic (T2D) mice and palmitate-induced MIN6 β-cells injury was observed. Fasting plasma glucose, triacylglycerol, total cholesterol, insulin levels and glucose tolerance test were evaluated. The expression of AMPK and JNK was detected in islet and MIN6 cells. KEY FINDINGS CK treatment (30 mg/kg) decreased fasting plasma glucose, triacylglycerol, total cholesterol, elevated plasma insulin levels and improved glucose tolerance in T2D mice. CK treatment attenuated islet cell apoptosis and caspase-3 activity accompanied by a decrease in AMPK and JNK activation. Meanwhile, CK treatment attenuated the palmitate-induced reduction in MIN6 β-cell viability, apoptosis and caspase-3 activity and activation of AMPK and JNK. The AMPK activator AICAR attenuated the CK-mediated inhibition of palmitate-induced apoptosis. SIGNIFICANCE These data suggest that CK treatment provides a beneficial anti-diabetic effect in mice with T2D and this protective effect may be mediated through prevention of β-cell apoptosis via inhibition of the AMPK-JNK pathway.
Collapse
Affiliation(s)
- Feng Ying Guan
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Jian Gu
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130018, China
| | - Ming Zhang
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Yingshi Ji
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Jing Li
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China
| | - Li Chen
- Department of Pharmacology, Key Laboratory of Pathobiology, Ministry of Education, Basic Medicine College, Jilin University, Changchun 130021, China.
| | - Grant M Hatch
- Department of Pharmacology & Therapeutics, University of Manitoba, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| |
Collapse
|
15
|
Fu XM, Liu SJ, Dan QQ, Wang YP, Lin N, Lv LY, Zou Y, Liu S, Zhou X, Wang TH. Combined Bone Mesenchymal Stem Cell and Olfactory Ensheathing Cell Transplantation Promotes Neural Repair Associated With CNTF Expression in Traumatic Brain-Injured Rats. Cell Transplant 2014; 24:1533-44. [PMID: 24612678 DOI: 10.3727/096368914x679345] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study examined the role of bone mesenchymal stem cell (BMSC) and olfactory ensheathing cell (OEC) cografting on neural function and underlying molecular mechanisms in acute stage of traumatic brain injury (TBI) rats. Eighty Sprague-Dawley (SD) female rats were randomly divided into five groups (n = 16 per category): sham operated group (Sham), weight-drop-induced TBI group (TBI), BMSC transplantation group (BMSC), OEC transplantation group (OEC), and cotransplantation group (CO). Eight rats were randomly selected from each group for behavioral and morphological assessment. Another category (n = 8 rats) was employed in the genetic expression detection. BMSCs were isolated from GFP mice and identified by CD44 antibody. OECs were isolated from the SD rats, identified by P75 antibody and labeled by Hoechst 33342. They were then transplanted into the surrounding tissue of the epicenter of TBI rats. The result of neurological severity scores revealed that BMSC or OEC transplantation alone and BMSC and OEC cografting significantly ameliorated the neurological deficits of TBI rats. Quantitative immunohistochemical analysis showed that graft-recipient animals possessed dramatically more neurons and regenerated axons and smaller amounts of astrocytes than controls 14 days posttransplantation (p < 0.05). However, the expressional level of ciliary neurotrophic factor significantly decreased in the cografting group as determined by RT-PCR (p < 0.05), and the Janus kinase/signal transducer and activator of transcription pathway was significantly activated at 7 days after cell transplantation (p < 0.05). This study is the first to report the role of cotransplantation of BMSCs and OECs in the therapy of TBI and explore its potential molecular mechanisms, therefore providing the important morphological and molecular biological evidence for the clinical application of BMSC and/or OEC transplantation in TBI.
Collapse
Affiliation(s)
- Xue-Mei Fu
- Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Protzek AOP, Costa-Júnior JM, Rezende LF, Santos GJ, Araújo TG, Vettorazzi JF, Ortis F, Carneiro EM, Rafacho A, Boschero AC. Augmented β-Cell Function and Mass in Glucocorticoid-Treated Rodents Are Associated with Increased Islet Ir-β /AKT/mTOR and Decreased AMPK/ACC and AS160 Signaling. Int J Endocrinol 2014; 2014:983453. [PMID: 25313308 PMCID: PMC4182854 DOI: 10.1155/2014/983453] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 04/25/2014] [Accepted: 08/17/2014] [Indexed: 12/31/2022] Open
Abstract
Glucocorticoid (GC) therapies may adversely cause insulin resistance (IR) that lead to a compensatory hyperinsulinemia due to insulin hypersecretion. The increased β-cell function is associated with increased insulin signaling that has the protein kinase B (AKT) substrate with 160 kDa (AS160) as an important downstream AKT effector. In muscle, both insulin and AMP-activated protein kinase (AMPK) signaling phosphorylate and inactivate AS160, which favors the glucose transporter (GLUT)-4 translocation to plasma membrane. Whether AS160 phosphorylation is modulated in islets from GC-treated subjects is unknown. For this, two animal models, Swiss mice and Wistar rats, were treated with dexamethasone (DEX) (1 mg/kg body weight) for 5 consecutive days. DEX treatment induced IR, hyperinsulinemia, and dyslipidemia in both species, but glucose intolerance and hyperglycemia only in rats. DEX treatment caused increased insulin secretion in response to glucose and augmented β-cell mass in both species that were associated with increased islet content and increased phosphorylation of the AS160 protein. Protein AKT phosphorylation, but not AMPK phosphorylation, was found significantly enhanced in islets from DEX-treated animals. We conclude that the augmented β-cell function developed in response to the GC-induced IR involves inhibition of the islet AS160 protein activity.
Collapse
Affiliation(s)
- André O. P. Protzek
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
| | - José M. Costa-Júnior
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
| | - Luiz F. Rezende
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
| | - Gustavo J. Santos
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
| | - Tiago Gomes Araújo
- School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jean F. Vettorazzi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
| | - Fernanda Ortis
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
- Department of Cell and Developmental Biology, Institute of Biomedical Science, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Everardo M. Carneiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
| | - Alex Rafacho
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianópolis, SC, Brazil
| | - Antonio C. Boschero
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), P.O. Box 6109, 13083-970 Campinas, SP, Brazil
- *Antonio C. Boschero:
| |
Collapse
|
17
|
Brandimarti P, Costa-Júnior JM, Ferreira SM, Protzek AO, Santos GJ, Carneiro EM, Boschero AC, Rezende LF. Cafeteria diet inhibits insulin clearance by reduced insulin-degrading enzyme expression and mRNA splicing. J Endocrinol 2013; 219:173-82. [PMID: 23959080 DOI: 10.1530/joe-13-0177] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Insulin clearance plays a major role in glucose homeostasis and insulin sensitivity in physiological and/or pathological conditions, such as obesity-induced type 2 diabetes as well as diet-induced obesity. The aim of the present work was to evaluate cafeteria diet-induced obesity-induced changes in insulin clearance and to explain the mechanisms underlying these possible changes. Female Swiss mice were fed either a standard chow diet (CTL) or a cafeteria diet (CAF) for 8 weeks, after which we performed glucose tolerance tests, insulin tolerance tests, insulin dynamics, and insulin clearance tests. We then isolated pancreatic islets for ex vivo glucose-stimulated insulin secretion as well as liver, gastrocnemius, visceral adipose tissue, and hypothalamus for subsequent protein analysis by western blot and determination of mRNA levels by real-time RT-PCR. The cafeteria diet induced insulin resistance, glucose intolerance, and increased insulin secretion and total insulin content. More importantly, mice that were fed a cafeteria diet demonstrated reduced insulin clearance and decay rate as well as reduced insulin-degrading enzyme (IDE) protein and mRNA levels in liver and skeletal muscle compared with the control animals. Furthermore, the cafeteria diet reduced IDE expression and alternative splicing in the liver and skeletal muscle of mice. In conclusion, a cafeteria diet impairs glucose homeostasis by reducing insulin sensitivity, but it also reduces insulin clearance by reducing IDE expression and alternative splicing in mouse liver; however, whether this mechanism contributes to the glucose intolerance or helps to ameliorate it remains unclear.
Collapse
Affiliation(s)
- P Brandimarti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), PO Box 6109, Campinas, SP, CEP 13083-865, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ramkumar KM, Vijayakumar RS, Vanitha P, Suganya N, Manjula C, Rajaguru P, Sivasubramanian S, Gunasekaran P. Protective effect of gallic acid on alloxan-induced oxidative stress and osmotic fragility in rats. Hum Exp Toxicol 2013; 33:638-49. [PMID: 24064907 DOI: 10.1177/0960327113504792] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the present study, we investigated the antioxidant effect of gallic acid (GA) on membrane lipid peroxidation and osmotic fragility in alloxan-induced diabetic Wistar rats. GA was administered orally at doses of 5, 10, and 20 mg/kg body weight for 45 days, after which liver and kidney tissues were analyzed for the degree of lipid peroxidation, reduced glutathione, and the activities of antioxidants such as catalase, superoxide dismutase, and glutathione peroxidase. Administration of GA to alloxan-induced diabetic rats reduced the blood glucose level with an increase in the level of insulin. Liver and kidney tissues from diabetic animals exhibited disturbances in antioxidant defense compared with normal rats. GA at a dose of 20 mg/kg b.w. showed a significant effect than that of the other doses. In addition, the results revealed that GA protected the integrity of erythrocyte membrane in diabetic rats as demonstrated by lower percentage of hemolysis and resistance to hydrogen peroxide-induced peroxidation. The anti-hyperglycemic activity of GA in alloxan-induced diabetic rats was also comparable with glibenclamide, a reference drug. These results suggest that GA could provide a beneficial effect on diabetes by decreasing oxidative stress-related diabetic complications.
Collapse
Affiliation(s)
- K M Ramkumar
- SRM Research Institute, SRM University, Kattankulathur, Chennai, Tamil Nadu, India
| | - R S Vijayakumar
- Institute of Biotechnology and Pharmaceutical Research, The National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - P Vanitha
- SRM Research Institute, SRM University, Kattankulathur, Chennai, Tamil Nadu, India
| | - N Suganya
- SRM Research Institute, SRM University, Kattankulathur, Chennai, Tamil Nadu, India
| | - C Manjula
- Department of Biotechnology, Anna University-BIT Campus, Tiruchirappalli, Tamil Nadu, India
| | - P Rajaguru
- Department of Biotechnology, Anna University-BIT Campus, Tiruchirappalli, Tamil Nadu, India
| | - S Sivasubramanian
- King Institute of Preventive Medicine and Research, Guindy, Chennai, Tamil Nadu, India
| | - P Gunasekaran
- King Institute of Preventive Medicine and Research, Guindy, Chennai, Tamil Nadu, India
| |
Collapse
|
19
|
Rezende LF, Santos GJ, Carneiro EM, Boschero AC. Ciliary neurotrophic factor protects mice against streptozotocin-induced type 1 diabetes through SOCS3: the role of STAT1/STAT3 ratio in β-cell death. J Biol Chem 2012; 287:41628-39. [PMID: 23038263 DOI: 10.1074/jbc.m112.358788] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Type 1 diabetes is characterized by a loss of islet β-cells. Ciliary neurotrophic factor (CNTF) protects pancreatic islets against cytokine-induced apoptosis. For this reason, we assessed whether CNTF protects mice against streptozotocin-induced diabetes (a model of type 1 diabetes) and the mechanism for this protection. WT and SOCS3 knockdown C57BL6 mice were treated for 5 days with citrate buffer or 0.1 mg/kg CNTF before receiving 80 mg/kg streptozotocin. Glycemia in non-fasted mice was measured weekly from days 0-28 after streptozotocin administration. Diabetes was defined as a blood glucose > 11.2 mmol/liter. Wild-type (WT) and SOCS3 knockdown MIN6 cells were cultured with CNTF, IL1β, or both. CNTF reduced diabetes incidence and islet apoptosis in WT but not in SOCS3kd mice. Likewise, CNTF inhibited apoptosis in WT but not in SOCS3kd MIN6 cells. CNTF increased STAT3 phosphorylation in WT and SOCS3kd mice and MIN6 cells but reduced STAT1 phosphorylation only in WT mice, in contrast to streptozotocin and IL1β. Moreover, CNTF reduced NFκB activation and required down-regulation of inducible NO synthase expression to exert its protective effects. In conclusion, CNTF protects mice against streptozotocin-induced diabetes by increasing pancreatic islet survival, and this protection depends on SOCS3. In addition, SOCS3 expression and β-cell fate are dependent on STAT1/STAT3 ratio.
Collapse
Affiliation(s)
- Luiz F Rezende
- Department of Structural and Functional Biology Institute of Biology State University of Campinas (UNICAMP), P.O. Box 6109, Campinas, Sao Paulo 13083-865, Brazil.
| | | | | | | |
Collapse
|
20
|
Rezende LF, Santos GJ, Santos-Silva JC, Carneiro EM, Boschero AC. Ciliary neurotrophic factor (CNTF) protects non-obese Swiss mice against type 2 diabetes by increasing beta cell mass and reducing insulin clearance. Diabetologia 2012; 55:1495-504. [PMID: 22349107 DOI: 10.1007/s00125-012-2493-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 01/19/2012] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Ciliary neurotrophic factor (CNTF) improves metabolic variables of obese animals with characteristics of type 2 diabetes, mainly by reducing insulin resistance. We evaluated whether CNTF was able to improve other metabolic variables in mouse models of type 2 diabetes, such as beta cell mass and insulin clearance, and whether CNTF has any effect on non-obese mice with characteristics of type 2 diabetes. METHODS Neonatal mice were treated with 0.1 mg/kg CNTF or citrate buffer via intraperitoneal injections, before injection of 250 mg/kg alloxan. HEPG2 cells were cultured for 3 days in the presence of citrate buffer, 1 nmol/l CNTF or 50 mmol/l alloxan or a combination of CNTF and alloxan. Twenty-one days after treatment, we determined body weight, epididymal fat weight, blood glucose, plasma insulin, NEFA, glucose tolerance, insulin resistance, insulin clearance and beta cell mass. Finally, we assessed insulin receptor and protein kinase B phosphorylation in peripheral organs, as well as insulin-degrading enzyme (IDE) protein production and alternative splicing in the liver and HEPG2 cells. RESULTS CNTF improved insulin sensitivity and beta cell mass, while reducing glucose-stimulated insulin secretion and insulin clearance in Swiss mice, improving glucose handling in a non-obese type 2 diabetes model. This effect was associated with lower IDE production and activity in liver cells. All these effects were observed even at 21 days after CNTF treatment. CONCLUSIONS/INTERPRETATION CNTF protection against type 2 diabetes is partially independent of the anti-obesity actions of CNTF, requiring a reduction in insulin clearance and increased beta cell mass, besides increased insulin sensitivity. Furthermore, knowledge of the long-term effects of CNTF expands its pharmacological relevance.
Collapse
Affiliation(s)
- L F Rezende
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, PO Box 6109, Campinas, SP CEP 13083-865, Brazil.
| | | | | | | | | |
Collapse
|