201
|
Peysepar E, Soltani N, Nematbakhsh M, Eshraghi-Jazi F, Talebi A. Gamma-aminobutyric acid aggravates nephrotoxicity induced by cisplatin in female rats. J Renal Inj Prev 2016; 5:188-92. [PMID: 27689121 PMCID: PMC5039987 DOI: 10.15171/jrip.2016.40] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/03/2016] [Indexed: 11/18/2022] Open
Abstract
Introduction: Cisplatin (CP) is a major antineoplastic drug for treatment of solid tumors. CP-induced nephrotoxicity may be gender-related. This is while gamma-aminobutyric acid (GABA) is an inhibitory neurotransmitter in the central nervous system that has renoprotective impacts on acute renal injury.
Objectives: This study was designed to investigate the protective role of GABA against CP-induced nephrotoxicity in male and female rats.
Materials and Methods: Sixty Wistar male and female rats were used in eight experimental groups. Both genders received GABA (50 μg/kg/day; i. p.) for 14 days and CP (2.5 mg/kg/day; i. p.) was added from day 8 to the end of the study, and they were compared with the control groups. At the end of the study, all animals were sacrificed and the serum levels of blood urea nitrogen (BUN), creatinine (Cr), nitrite, malondialdehyde (MDA), and magnesium (Mg) were measured. The kidney tissue damage was also determined via staining.
Results: CP significantly increased the serum levels of Cr and BUN, kidney weight, and kidney tissue damage score in both genders (P<0.05). GABA did not attenuate these markers in males; even these biomarkers were intensified in females. Serum level of Mg, and testis and uterus weights did not alter in the groups. However, the groups were significantly different in terms of nitrite and MDA levels.
Conclusion: It seems that GABA did not improve nephrotoxicity induced by CP-treated rats, and it exacerbated renal damage in female rats.
Collapse
Affiliation(s)
- Elham Peysepar
- Department of Physiology, Hormozgan University of Medical Sciences, Bandar Abbas, Iran ; Water & Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nepton Soltani
- Department of Physiology, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mehdi Nematbakhsh
- Water & Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran ; Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran ; Isfahan MN Institute of Basic & Applied Sciences Research, Isfahan, Iran
| | - Fatemeh Eshraghi-Jazi
- Water & Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ardeshir Talebi
- Water & Electrolytes Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
202
|
Urrutia M, Fernández S, González M, Vilches R, Rojas P, Vásquez M, Kurte M, Vega-Letter AM, Carrión F, Figueroa F, Rojas P, Irarrázabal C, Fuentealba RA. Overexpression of Glutamate Decarboxylase in Mesenchymal Stem Cells Enhances Their Immunosuppressive Properties and Increases GABA and Nitric Oxide Levels. PLoS One 2016; 11:e0163735. [PMID: 27662193 PMCID: PMC5035029 DOI: 10.1371/journal.pone.0163735] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
The neurotransmitter GABA has been recently identified as a potent immunosuppressive agent that targets both innate and adaptive immune systems and prevents disease progression of several autoimmunity models. Mesenchymal stem cells (MSCs) are self-renewing progenitor cells that differentiate into various cell types under specific conditions, including neurons. In addition, MSC possess strong immunosuppressive capabilities. Upon cytokine priming, undifferentiated MSC suppress T-cell proliferation via cell-to-cell contact mechanisms and the secretion of soluble factors like nitric oxide, prostaglandin E2 and IDO. Although MSC and MSC-derived neuron-like cells express some GABAergic markers in vitro, the role for GABAergic signaling in MSC-mediated immunosuppression remains completely unexplored. Here, we demonstrate that pro-inflammatory cytokines selectively regulate GAD-67 expression in murine bone marrow-MSC. However, expression of GAD-65 is required for maximal GABA release by MSC. Gain of function experiments using GAD-67 and GAD-65 co-expression demonstrates that GAD increases immunosuppressive function in the absence of pro-inflammatory licensing. Moreover, GAD expression in MSC evokes an increase in both GABA and NO levels in the supernatants of co-cultured MSC with activated splenocytes. Notably, the increase in NO levels by GAD expression was not observed in cultures of isolated MSC expressing GAD, suggesting crosstalk between these two pathways in the setting of immunosuppression. These results indicate that GAD expression increases MSC-mediated immunosuppression via secretion of immunosuppressive agents. Our findings may help reconsider GABAergic activation in MSC for immunological disorders.
Collapse
Affiliation(s)
- Mariana Urrutia
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Sebastián Fernández
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Marisol González
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rodrigo Vilches
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Pablo Rojas
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Manuel Vásquez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Mónica Kurte
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Flavio Carrión
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Fernando Figueroa
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Patricio Rojas
- Departamento de Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Carlos Irarrázabal
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Rodrigo A. Fuentealba
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- * E-mail:
| |
Collapse
|
203
|
Affiliation(s)
- Linda V Thomas
- Yakult UK Limited; Odyssey Business Park West End Road South Ruislip London HA4 6QQ UK
| |
Collapse
|
204
|
Chandravanshi B, Bhonde R. Small molecules exert anti-apoptotic effect and reduce oxidative stress augmenting insulin secretion in stem cells engineered islets against hypoxia. Eur J Pharmacol 2016; 791:424-432. [PMID: 27614129 DOI: 10.1016/j.ejphar.2016.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 01/09/2023]
Abstract
Transplantation of pancreatic islets is the most reliable treatment for Type 1 diabetes. However cell death mediated by hypoxia is considered as one of the main difficulties hindering success in islet transplantation. The aim of our experiment was to investigate the role of small molecules in survival of Islet like cell aggregates (ICAs) engineered from umbilical cord matrix under oxygen deprived condition (<5% O2). ICAs were analyzed for cell death via fluoroscein diacetate/propidium iodide (FDA/PI) staining, estimation of Caspase 3 and free radical release in presence and absence of small molecules. The samples were also analyzed for the presence of hypoxia inducible factor 1α (HIF1α) at both transcriptional and translational level. The addition of small molecules showed profound defensive effect on ICAs under hypoxic environment as evidenced by their viability and insulin secretion compared to untreated ICAs. The combinations of Eicosapentaenoic acid (EPA), Docosahexaenoic acid(DHA) and metformin and EPA, DHAandγ amino butyric acid (GABA) acted as anti-apoptotic agents for human ICAs when exposed to 1% O2 for 48h. The combinations of the small molecules reduced the total reactive oxygen species and malonaldehyde (MDA) levels and enhanced the production of glutathione peroxidise (GPx) enzyme under hypoxic conditions. Finally the increase in HIF1α at both protein and gene level confirmed the defensive effect of the additives in hypoxia. These results suggest that the combination of small molecules maintained the viability and functionality of the ICAs in hypoxia by up-regulating HIF1α expression and down regulating the Caspase 3 activity.
Collapse
Affiliation(s)
- Bhawna Chandravanshi
- School of Regenerative Medicine, Manipal University, GKVK Post, Bellary Road, Allalasandra, Yelahanka, 560065 Bangalore, India
| | - Ramesh Bhonde
- School of Regenerative Medicine, Manipal University, GKVK Post, Bellary Road, Allalasandra, Yelahanka, 560065 Bangalore, India.
| |
Collapse
|
205
|
Phelps EA, Cianciaruso C, Michael IP, Pasquier M, Kanaani J, Nano R, Lavallard V, Billestrup N, Hubbell JA, Baekkeskov S. Aberrant Accumulation of the Diabetes Autoantigen GAD65 in Golgi Membranes in Conditions of ER Stress and Autoimmunity. Diabetes 2016; 65:2686-99. [PMID: 27284108 PMCID: PMC5001175 DOI: 10.2337/db16-0180] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/27/2016] [Indexed: 02/06/2023]
Abstract
Pancreatic islet β-cells are particularly susceptible to endoplasmic reticulum (ER) stress, which is implicated in β-cell dysfunction and loss during the pathogenesis of type 1 diabetes (T1D). The peripheral membrane protein GAD65 is an autoantigen in human T1D. GAD65 synthesizes γ-aminobutyric acid, an important autocrine and paracrine signaling molecule and a survival factor in islets. We show that ER stress in primary β-cells perturbs the palmitoylation cycle controlling GAD65 endomembrane distribution, resulting in aberrant accumulation of the palmitoylated form in trans-Golgi membranes. The palmitoylated form has heightened immunogenicity, exhibiting increased uptake by antigen-presenting cells and T-cell stimulation compared with the nonpalmitoylated form. Similar accumulation of GAD65 in Golgi membranes is observed in human β-cells in pancreatic sections from GAD65 autoantibody-positive individuals who have not yet progressed to clinical onset of T1D and from patients with T1D with residual β-cell mass and ongoing T-cell infiltration of islets. We propose that aberrant accumulation of immunogenic GAD65 in Golgi membranes facilitates inappropriate presentation to the immune system after release from stressed and/or damaged β-cells, triggering autoimmunity.
Collapse
Affiliation(s)
- Edward A Phelps
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Chiara Cianciaruso
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Iacovos P Michael
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Miriella Pasquier
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jamil Kanaani
- Departments of Medicine, Microbiology and Immunology and Diabetes Center, University of California San Francisco, San Francisco, CA
| | - Rita Nano
- Diabetes Research Institute, IRCCS, Pancreatic Islet Processing Facility, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Faculty of Medicine, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Nils Billestrup
- Section of Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeffrey A Hubbell
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland Institute for Molecular Engineering, University of Chicago, Chicago, IL
| | - Steinunn Baekkeskov
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland Graduate Program in Biotechnology and Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland Departments of Medicine, Microbiology and Immunology and Diabetes Center, University of California San Francisco, San Francisco, CA
| |
Collapse
|
206
|
Affiliation(s)
- Ralf P. Brandes
- From the Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
207
|
Liu W, Yang Y, Liu Y, Lu X, Guo S, Wu M, Wang M, Yan L, Wang Q, Zhao X, Tong X, Hu J, Li Y, Hu R, Stanton RC, Zhang Z. Exogenous kallikrein protects against diabetic nephropathy. Kidney Int 2016; 90:1023-1036. [PMID: 27546607 DOI: 10.1016/j.kint.2016.06.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 06/13/2016] [Accepted: 06/16/2016] [Indexed: 01/06/2023]
Abstract
The kallikrein-kinin system has been shown to be involved in the development of diabetic nephropathy, but specific mechanisms are not fully understood. Here, we determined the renal-protective role of exogenous pancreatic kallikrein in diabetic mice and studied potential mechanisms in db/db type 2 diabetic and streptozotocin-induced type 1 diabetic mice. After the onset of diabetes, mice were treated with either pancreatic kallikrein (db/db+kallikrein, streptozotocin+kallikrein) or saline (db/db+saline, streptozotocin+saline) for 16 weeks, while another group of streptozotocin-induced diabetic mice received the same treatment after onset of albuminuria (streptozotocin'+kallikrein, streptozotocin'+saline). Db/m littermates or wild type mice were used as non-diabetic controls. Pancreatic kallikrein had no effects on body weight, blood glucose and blood pressure, but significantly reduced albuminuria among all three groups. Pathological analysis showed that exogenous kallikrein decreased the thickness of the glomerular basement membrane, protected against the effacement of foot process, the loss of endothelial fenestrae, and prevented the loss of podocytes in diabetic mice. Renal fibrosis, inflammation and oxidative stress were reduced in kallikrein-treated mice compared to diabetic controls. The expression of kininogen1, tissue kallikrein, kinin B1 and B2 receptors were all increased in the kallikrein-treated compared to saline-treated mice. Thus, exogenous pancreatic kallikrein both prevented and ameliorated diabetic nephropathy, which may be mediated by activating the kallikrein-kinin system.
Collapse
Affiliation(s)
- Wenjuan Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yemei Liu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China; Department of Endocrinology, The Second People's Hospital, Wuhu, Anhui, China
| | - Xiaolan Lu
- Department of Endocrinology, High-tech District People's Hospital, Suzhou, Jiangsu, China
| | - Shizhe Guo
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Meng Wu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Meng Wang
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Linling Yan
- Department of Endocrinology, The First People's Hospital of Taichang, Jiangsu, China
| | - Qinghua Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetology, Fudan University, Shanghai, China; Division of Endocrinology and Metabolism, Keenan Research Centre at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Xiaolong Zhao
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Xian Tong
- Jiangsu (Qianhong) Engineering Research Center for Innovative Biological Drugs, Changzhou, Jiangsu, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Yiming Li
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetology, Fudan University, Shanghai, China
| | - Renming Hu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetology, Fudan University, Shanghai, China
| | - Robert C Stanton
- Renal Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China; Institute of Endocrinology and Diabetology, Fudan University, Shanghai, China.
| |
Collapse
|
208
|
Gamma Amino Butyric Acid Attenuates Brain Oxidative Damage Associated with Insulin Alteration in Streptozotocin-Treated Rats. Indian J Clin Biochem 2016; 32:207-213. [PMID: 28428696 DOI: 10.1007/s12291-016-0597-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/23/2016] [Indexed: 10/21/2022]
Abstract
The aim of the current study was to evaluate the role of γ-amino butyric acid (GABA) in insulin disturbance and hyperglycemia associated with brain oxidative damage in streptozotocin-treated rats. Streptozotocin (STZ) was administered to male albino rats as a single intraperitoneal dose (60 mg/kg body weight). GABA (200 mg/Kg body weight/day) was administered daily via gavages during 3 weeks to STZ-treated-rats. Male albino rats Sprague-Dawley (10 ± 2 weeks old; 120 ± 10 g body weight) were divided into 4 groups of 6 rats and treated in parallel. (1) Control group: received distilled water, (2) GABA group: received GABA, (3) STZ group: STZ-treated rats received distilled water, (4) STZ + GABA group: STZ-treated rats received GABA. Rats were sacrificed after a fasting period of 12 h next last dose of GABA. The results obtained showed that STZ-treatment produced hyperglycemia and insulin deficiency (similar to type1 Diabetes). These changes were associated with oxidative damage in brain tissue and notified by significant decreases of superoxide dismutase and catalase activities in parallel to significant increases of malondialdehyde and advanced oxidation protein products levels. The histopathology reports also revealed that STZ-treatment produced degeneration of pancreatic cells. The administration of GABA to STZ-treated rats preserved pancreatic tissue with improved insulin secretion, improved glucose level and minimized oxidative stress in brain tissues. It could be concluded that GABA might protect the brain from oxidative stress and preserve pancreas tissues with adjusting glucose and insulin levels in Diabetic rats and might decrease the risk of neurodegenerative disease in diabetes.
Collapse
|
209
|
Kim MJ, Lim JS, Yang SA. Component Analysis and Anti-Proliferative Effects of Ethanol Extracts of Fruits, Leaves, and Stems from Elaeagnus umbellata in HepG2 Cells. ACTA ACUST UNITED AC 2016. [DOI: 10.3746/jkfn.2016.45.6.828] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
210
|
Sen S, Roy S, Bandyopadhyay G, Scott B, Xiao D, Ramadoss S, Mahata SK, Chaudhuri G. γ-Aminobutyric Acid Is Synthesized and Released by the Endothelium: Potential Implications. Circ Res 2016; 119:621-34. [PMID: 27354210 DOI: 10.1161/circresaha.116.308645] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/28/2016] [Indexed: 11/16/2022]
Abstract
RATIONALE Gamma aminobutyric acid (GABA), a neurotransmitter of the central nervous system, is found in the systemic circulation of humans at a concentration between 0.5 and 3 μmol/L. However, the potential source of circulating GABA and its significance on the vascular system remains unknown. We hypothesized that endothelial cells (ECs) may synthesize and release GABA to modulate some functions in the EC and after its release into the circulation. OBJECTIVE To assess whether GABA is synthesized and released by the EC and its potential functions. METHODS AND RESULTS Utilizing the human umbilical vein ECs and aortic ECs, we demonstrated for the first time that ECs synthesize and release GABA from [1-(14)C]glutamate. Localization of GABA and the presence of the GABA-synthesizing enzyme, glutamic acid decarboxylase in EC were confirmed by immunostaining and immunoblot analysis, respectively. The presence of GABA was further confirmed by immunohistochemistry in the EC lining the human coronary vessel. EC-derived GABA regulated the key mechanisms of ATP synthesis, fatty acid, and pyruvate oxidation in EC. GABA protected EC by inhibiting the reactive oxygen species generation and prevented monocyte adhesion by attenuating vascular cell adhesion molecule -1 and monocyte chemoattractant protein-1 expressions. GABA had no relaxing effect on rat aortic rings. GABA exhibited a dose-dependent fall in blood pressure. However, the fall in BP was abolished after pretreatment with pentolinium. CONCLUSIONS Our findings indicate novel potential functions of endothelium-derived GABA.
Collapse
Affiliation(s)
- Suvajit Sen
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.).
| | - Sohini Roy
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.)
| | - Gautam Bandyopadhyay
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.)
| | - Bari Scott
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.)
| | - Daliao Xiao
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.)
| | - Sivakumar Ramadoss
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.)
| | - Sushil K Mahata
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.)
| | - Gautam Chaudhuri
- From the Department of Obstetrics and Gynecology (S.S., S.R., B.S., S.R., G.C.) and Department of Molecular and Medical Pharmacology (G.C.) David Geffen School of Medicine at University of California at Los Angeles; Jonsson Comprehensive Cancer Center, Los Angeles, CA (S.S., G.C.); Department of Medicine, University of California San Diego, VA San Diego Health Care System (G.B., S.K.M.); and Division of Pharmacology, Department of Basic Sciences, Center for Perinatal Biology, Loma Linda University School of Medicine, CA (D.X.).
| |
Collapse
|
211
|
Shen ML, Wang CH, Chen RYT, Zhou N, Kao ST, Wu DC. Luteolin inhibits GABAA receptors in HEK cells and brain slices. Sci Rep 2016; 6:27695. [PMID: 27292079 PMCID: PMC4904371 DOI: 10.1038/srep27695] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/24/2016] [Indexed: 11/23/2022] Open
Abstract
Modulation of the A type γ-aminobutyric acid receptors (GABAAR) is one of the major drug targets for neurological and psychological diseases. The natural flavonoid compound luteolin (2-(3,4-Dihydroxyphenyl)- 5,7-dihydroxy-4-chromenone) has been reported to have antidepressant, antinociceptive, and anxiolytic-like effects, which possibly involve the mechanisms of modulating GABA signaling. However, as yet detailed studies of the pharmacological effects of luteolin are still lacking, we investigated the effects of luteolin on recombinant and endogenous GABAAR-mediated current responses by electrophysiological approaches. Our results showed that luteolin inhibited GABA-mediated currents and slowed the activation kinetics of recombinant α1β2, α1β2γ2, α5β2, and α5β2γ2 receptors with different degrees of potency and efficacy. The modulatory effect of luteolin was likely dependent on the subunit composition of the receptor complex: the αβ receptors were more sensitive than the αβγ receptors. In hippocampal pyramidal neurons, luteolin significantly reduced the amplitude and slowed the rise time of miniature inhibitory postsynaptic currents (mIPSCs). However, GABAAR-mediated tonic currents were not significantly influenced by luteolin. These data suggested that luteolin has negative modulatory effects on both recombinant and endogenous GABAARs and inhibits phasic rather than tonic inhibition in hippocampus.
Collapse
Affiliation(s)
- Mei-Lin Shen
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chen-Hung Wang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Rita Yu-Tzu Chen
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Ning Zhou
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Shung-Te Kao
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Dong Chuan Wu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
212
|
Rana S, Kumar S, Rathore N, Padwad Y, Bhushana S. Nutrigenomics and its Impact on Life Style Associated Metabolic Diseases. Curr Genomics 2016; 17:261-78. [PMID: 27252592 PMCID: PMC4869012 DOI: 10.2174/1389202917666160202220422] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 02/07/2023] Open
Abstract
Post-human genome revelation observes the emergence of 'Nutigenomics' as one of the exciting scientific advancement influencing mankind around the world. Food or more precisely 'nutrition' has the major impact in defining the cause-response interaction between nutrient (diet) and human health. In addition to substantial understanding of nutrition-human-health interaction, bases of 'nutrigenomic' development foster on advent in transcriptomics, genomics, proteomics and metabolomics as well as insight into food as health supplement. Interaction of selected nutrient with associated genes in specific organ or tissue necessary to comprehend that how individual's genetic makeup (DNA transcribed into mRNA and then to proteins) respond to particular nutrient. It provided new opportunities to incorporate natural bioactive compounds into food for specific group of people with similar genotype. As inception of diabetes associated with change in gene expression of, not limited to, protein kinase B, insulin receptor, duodenal homeobox and glucokinase, thus, targeting such proteins by modifying or improving the nutritional availability or uptake may help to devise novel food, supplements, or nutraceuticals. In this article, various aspects of R&D in nutrigenomics are reviewed to ascertain its impact on human health, especially with life-style associated diseases.
Collapse
Affiliation(s)
- Shalika Rana
- Department of Biotechnology, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176 061 (HP), India
- Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology,
Palampur176 061 (HP), India
| | - Shiv Kumar
- Pharmacology and Toxicology Lab, Department of Food Nutraceuticals and Quality
Control, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176 061 (HP), India
- Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology,
Palampur176 061 (HP), India
| | - Nikita Rathore
- Department of Biotechnology, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176 061 (HP), India
| | - Yogendra Padwad
- Pharmacology and Toxicology Lab, Department of Food Nutraceuticals and Quality
Control, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176 061 (HP), India
- Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology,
Palampur176 061 (HP), India
| | - Shashi Bhushana
- Department of Biotechnology, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176 061 (HP), India
- Academy of Scientific and Innovative Research, CSIR-Institute of Himalayan Bioresource Technology,
Palampur176 061 (HP), India
| |
Collapse
|
213
|
He S, Zhang Y, Wang D, Tao K, Zhang S, Wei L, Chen Q. Rapamycin/GABA combination treatment ameliorates diabetes in NOD mice. Mol Immunol 2016; 73:130-7. [PMID: 27082922 DOI: 10.1016/j.molimm.2016.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/14/2016] [Accepted: 01/22/2016] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes (T1D) is one of the most common autoimmune diseases, affecting nearly 20 million people worldwide. The pathogenesis of T1D is characterized by the progressive destruction of insulin-producing pancreatic β-cells by autoreactive T cells. The significant role of immunomodulation in preserving residual insulin-producing β-cells in newly diagnosed T1D has not been confirmed yet. However, a combination of treatments acting via distinct mechanisms is widely considered to be the most promising future therapeutic approach in this respect. Rapamycin and gamma-aminobutyric acid (GABA) administration alone showed no effects on late-stage disease. By contrast, combined rapamycin/GABA treatment effectively suppressed autoimmune responses to islet cells and improved islet function in recent-onset diabetes. In particular, after the onset of hyperglycemia, those treated with the rapamycin/GABA combination showed significant amelioration of diabetes amelioration compared to those treated with either rapamycin or GABA alone. This protective effect of the rapamycin/GABA combination treatment in nonobese diabetic (NOD) mice was exerted through two distinct mechanisms. Rapamycin induced regulatory T cells and consequently suppressed targeted autoimmune responses to islet cells, which may be relevant to the reduced insulitis observed in rapamycin-treated NOD mice. By contrast, treatment with GABA improved islet function in diabetic NOD mice. We believe that our observations are highly relevant to establishing clinical strategies for the prevention and treatment of T1D in future.
Collapse
Affiliation(s)
- Sirong He
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Yi Zhang
- Department of Hepatobiliopancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Dan Wang
- Department of Respiration Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Kun Tao
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Shuang Zhang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Lingling Wei
- Center for Cell Transplantation (Seventh Unit of General Surgery Department), Institute of Organ Transplantation, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu 610072,PR China
| | - Quan Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
214
|
Ando A, Nakamura T. Prevention of GABA reduction during dough fermentation using a baker's yeast dal81 mutant. J Biosci Bioeng 2016; 122:441-5. [PMID: 27056577 DOI: 10.1016/j.jbiosc.2016.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 02/01/2016] [Accepted: 03/11/2016] [Indexed: 10/22/2022]
Abstract
γ-Aminobutyric acid (GABA) is consumed by yeasts during fermentation. To prevent GABA reduction in bread dough, a baker's yeast mutant AY77 deficient in GABA assimilation was characterized and utilized for wheat dough fermentation. An amber mutation in the DAL81 gene, which codes for a positive regulator of multiple nitrogen degradation pathways, was found in the AY77 strain. The qPCR analyses of genes involved in nitrogen utilization showed that transcriptional levels of the UGA1 and DUR3 genes encoding GABA transaminase and urea transporter, respectively, are severely decreased in the AY77 cells. The AY77 strain cultivated by fed-batch culture using cane molasses exhibited inferior gas production during dough fermentation compared to that of wild-type strain AY13. However, when fed with molasses containing 0.5% ammonium sulfate, the mutant strain exhibited gas production comparable to that of the AY13 strain. In contrast to the AY13 strain, which completely consumed GABA in dough within 5 h, the AY77 strain consumed no GABA under either culture condition. Dough fermentation with the dal81 mutant strain should be useful for suppression of GABA reduction in breads.
Collapse
Affiliation(s)
- Akira Ando
- NARO Institute of Vegetable and Tea Science, 360 Kusawa, Ano, Tsu, Mie 514-2392 Japan
| | - Toshihide Nakamura
- Yeast Laboratory, National Food Research Institute, National Agriculture and Food Research Organization (NARO), 2-1-12 Kannondai, Tsukuba, Ibaraki 305-8642 Japan.
| |
Collapse
|
215
|
Marques TM, Patterson E, Wall R, O'Sullivan O, Fitzgerald GF, Cotter PD, Dinan TG, Cryan JF, Ross RP, Stanton C. Influence of GABA and GABA-producing Lactobacillus brevis DPC 6108 on the development of diabetes in a streptozotocin rat model. Benef Microbes 2016; 7:409-20. [PMID: 27013462 DOI: 10.3920/bm2015.0154] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of this study was to investigate if dietary administration of γ-aminobutyric acid (GABA)-producing Lactobacillus brevis DPC 6108 and pure GABA exert protective effects against the development of diabetes in streptozotocin (STZ)-induced diabetic Sprague Dawley rats. In a first experiment, healthy rats were divided in 3 groups (n=10/group) receiving placebo, 2.6 mg/kg body weight (bw) pure GABA or L. brevis DPC 6108 (~10(9)microorganisms). In a second experiment, rats (n=15/group) were randomised to five groups and four of these received an injection of STZ to induce type 1 diabetes. Diabetic and non-diabetic controls received placebo [4% (w/v) yeast extract in dH2O], while the other three diabetic groups received one of the following dietary supplements: 2.6 mg/kg bw GABA (low GABA), 200 mg/kg bw GABA (high GABA) or ~10(9) L. brevis DPC 6108. L. brevis DPC 6108 supplementation was associated with increased serum insulin levels (P<0.05), but did not alter other metabolic markers in healthy rats. Diabetes induced by STZ injection decreased body weight (P<0.05), increased intestinal length (P<0.05) and stimulated water and food intake. Insulin was decreased (P<0.05), whereas glucose was increased (P<0.001) in all diabetic groups, compared with non-diabetic controls. A decrease (P<0.01) in glucose levels was observed in diabetic rats receiving L. brevis DPC 6108, compared with diabetic-controls. Both the composition and diversity of the intestinal microbiota were affected by diabetes. Microbial diversity in diabetic rats supplemented with low GABA was not reduced (P>0.05), compared with non-diabetic controls while all other diabetic groups displayed reduced diversity (P<0.05). L. brevis DPC 6108 attenuated hyperglycaemia induced by diabetes but additional studies are needed to understand the mechanisms involved in this reduction.
Collapse
Affiliation(s)
- T M Marques
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,2 Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.,3 School of Microbiology, University College Cork, Cork, Ireland
| | - E Patterson
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,2 Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland.,3 School of Microbiology, University College Cork, Cork, Ireland
| | - R Wall
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,2 Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - O O'Sullivan
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,2 Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - G F Fitzgerald
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,3 School of Microbiology, University College Cork, Cork, Ireland
| | - P D Cotter
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,2 Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - T G Dinan
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,4 Department of Psychiatry and Neurobehavioural Science, Biosciences Institute, University College Cork, Cork, Ireland
| | - J F Cryan
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland
| | - R P Ross
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland
| | - C Stanton
- 1 APC Microbiome Institute, University College Cork, Cork, Ireland.,2 Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| |
Collapse
|
216
|
Otter S, Lammert E. Exciting Times for Pancreatic Islets: Glutamate Signaling in Endocrine Cells. Trends Endocrinol Metab 2016; 27:177-188. [PMID: 26740469 DOI: 10.1016/j.tem.2015.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 01/14/2023]
Abstract
Glutamate represents a key excitatory neurotransmitter in the central nervous system, and also modulates the function and viability of endocrine cells in pancreatic islets. In insulin-secreting beta cells, glutamate acts as an intracellular messenger, and its transport into secretory granules promotes glucose- and incretin-stimulated insulin secretion. Mitochondrial degradation of glutamate also contributes to insulin release when glutamate dehydrogenase is allosterically activated. It also signals extracellularly via glutamate receptors (AMPA and NMDA receptors) to modulate glucagon, insulin and somatostatin secretion, and islet cell survival. Its degradation products, GABA and γ-hydroxybutyrate, are released and also influence islet cell behavior. Thus, islet glutamate receptors, such as the NMDA receptors, might serve as possible drug targets to develop new medications for adjunct treatment of diabetes.
Collapse
Affiliation(s)
- Silke Otter
- Institute of Metabolic Physiology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, and German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, and German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany.
| |
Collapse
|
217
|
Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF, Stanton C. Gut microbiota, obesity and diabetes. Postgrad Med J 2016; 92:286-300. [PMID: 26912499 DOI: 10.1136/postgradmedj-2015-133285] [Citation(s) in RCA: 345] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
The central role of the intestinal microbiota in the progression and, equally, prevention of metabolic dysfunction is becoming abundantly apparent. The symbiotic relationship between intestinal microbiota and host ensures appropriate development of the metabolic system in humans. However, disturbances in composition and, in turn, functionality of the intestinal microbiota can disrupt gut barrier function, a trip switch for metabolic endotoxemia. This low-grade chronic inflammation, brought about by the influx of inflammatory bacterial fragments into circulation through a malfunctioning gut barrier, has considerable knock-on effects for host adiposity and insulin resistance. Conversely, recent evidence suggests that there are certain bacterial species that may interact with host metabolism through metabolite-mediated stimulation of enteric hormones and other systems outside of the gastrointestinal tract, such as the endocannabinoid system. When the abundance of these keystone species begins to decline, we see a collapse of the symbiosis, reflected in a deterioration of host metabolic health. This review will investigate the intricate axis between the microbiota and host metabolism, while also addressing the promising and novel field of probiotics as metabolic therapies.
Collapse
Affiliation(s)
- Elaine Patterson
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Paul M Ryan
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Co. Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Psychiatry and Neurobehavioural Science, University College Cork, Co. Cork, Ireland
| | - R Paul Ross
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland College of Science, Engineering and Food Science, University College Cork, Co. Cork, Ireland
| | - Gerald F Fitzgerald
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
218
|
Kharazmi F, Soltani N, Rezaei S, Keshavarz M, Farsi L. Role of GABAB receptor and L-Arg in GABA-induced vasorelaxation in non-diabetic and streptozotocin-induced diabetic rat vessels. IRANIAN BIOMEDICAL JOURNAL 2016; 19:91-5. [PMID: 25864813 PMCID: PMC4412919 DOI: 10.6091/ibj.1461.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background: Hypertension is considered an independent risk factor for cardiovascular mortality in diabetic patients. The present study was designed to determine the role of gamma amino butyric acid B (GABAB) receptor and L-arginine (L-Arg) in GABA-induced vasorelaxation in normal and streptozotocin-induced diabetic rat vessels. Methods: Diabetes was induced by a single i.p. injection of streptozotocin (STZ, 60 mg/kg). Eight weeks later, superior mesenteric arteries of all groups were isolated and perfused according to the McGregor method. Results: Baseline perfusion pressure of STZ diabetic rats was significantly higher than non-diabetic rats in both intact and denuded endothelium. In the presence of faclofen, a selective GABAB receptor blocker, GABA-induced relaxation in intact and denuded endothelium mesenteric beds of STZ diabetic rats was suppressed, but this response in non-diabetic rats was not suppressed. Our results showed that in the presence of L-Arg, a nitric oxide precursor, GABA induced vasorelaxation in both diabetic and non-diabetic vessels. Conclusion: From the results of this study, it may be concluded that the vasorelaxatory effect of GABA in diabetic vessel is mediated by the GABAB receptor and nitric oxide, but it seems that in non-diabetic vessel GABAB receptor does not play any role in GABA-induced vasorelaxation, but nitric oxide induced GABA relaxation in non-diabetic vessel.
Collapse
Affiliation(s)
- Fatemah Kharazmi
- Dept. of Physiology, Faculty of Medicine, Molecular Medicine Research Center, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Nepton Soltani
- Dept. of Physiology, Faculty of Medicine, Molecular Medicine Research Center, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Sana Rezaei
- Dept. of Physiology, Faculty of Medicine, Molecular Medicine Research Center, Hormozgan University of Medical Science, Bandar Abbas, Iran
| | - Mansoor Keshavarz
- Dept. of Physiology, Faculty of Medicine, Tehran University
of Medical Science, Tehran, Iran
| | - Leila Farsi
- Dept. of Physiology, Faculty of Medicine, Tehran University
of Medical Science, Tehran, Iran
| |
Collapse
|
219
|
γ-Aminobutyric acid-rich yogurt fermented by Streptococcus salivarius subsp. thermophiles fmb5 apprars to have anti-diabetic effect on streptozotocin-induced diabetic mice. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.10.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
220
|
Challenges and issues with streptozotocin-induced diabetes - A clinically relevant animal model to understand the diabetes pathogenesis and evaluate therapeutics. Chem Biol Interact 2015; 244:49-63. [PMID: 26656244 DOI: 10.1016/j.cbi.2015.11.032] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023]
Abstract
Streptozotocin (STZ) has been extensively used over the last three decades to induce diabetes in various animal species and to help screen for hypoglycemic drugs. STZ induces clinical features in animals that resemble those associated with diabetes in humans. For this reason STZ treated animals have been used to study diabetogenic mechanisms and for preclinical evaluation of novel antidiabetic therapies. However, the physiochemical characteristics and associated toxicities of STZ are still major obstacles for researchers using STZ treated animals to investigate diabetes. Another major challenges in STZ-induced diabetes are sustaining uniformity, suitability, reproducibility and induction of diabetes with minimal animal lethality. Lack of appropriate use of STZ was found to be associated with increased mortality and animal suffering. During STZ use in animals, attention should be paid to several factors such as method of preparation of STZ, stability, suitable dose, route of administration, diet regimen, animal species with respect to age, body weight, gender and the target blood glucose level used to represent hyperglycemia. Therefore, protocol for STZ-induced diabetes in experimental animals must be meticulously planned. This review highlights specific skills and strategies involved in the execution of STZ-induced diabetes model. The present review aims to provide insight into diabetogenic mechanisms of STZ, specific toxicity of STZ with its significance and factors responsible for variations in diabetogenic effects of STZ. Further this review also addresses ways to minimize STZ-induced mortality, suggests methods to improve STZ-based experimental models and best utilize them for experimental studies purported to understand diabetes pathogenesis and preclinical evaluation of drugs.
Collapse
|
221
|
Li J, Zhang Z, Liu X, Wang Y, Mao F, Mao J, Lu X, Jiang D, Wan Y, Lv JY, Cao G, Zhang J, Zhao N, Atkinson M, Greiner DL, Prud'homme GJ, Jiao Z, Li Y, Wang Q. Study of GABA in Healthy Volunteers: Pharmacokinetics and Pharmacodynamics. Front Pharmacol 2015; 6:260. [PMID: 26617516 PMCID: PMC4639630 DOI: 10.3389/fphar.2015.00260] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/20/2015] [Indexed: 01/13/2023] Open
Abstract
Preclinical studies show that GABA exerts anti-diabetic effects in rodent models of type 1 diabetes. Because little is known about its absorption and effects in humans, we investigated the pharmacokinetics and pharmacodynamics of GABA in healthy volunteers. Twelve subjects were subjected to an open-labeled, three-period trial involving sequential oral administration of placebo, 2 g GABA once, and 2 g GABA three times/day for 7 days, with a 7-day washout between each period. GABA was rapidly absorbed (Tmax: 0.5 ~ 1 h) with the half-life (t1/2) of 5 h. No accumulation was observed after repeated oral GABA administration for 7 days. Remarkably, GABA significantly increased circulating insulin levels in the subjects under either fasting (1.6-fold, single dose; 2.0-fold, repeated dose; p < 0.01) or fed conditions (1.4-fold, single dose; 1.6-fold, repeated dose; p < 0.01). GABA also increased glucagon levels only under fasting conditions (1.3-fold, single dose, p < 0.05; 1.5-fold, repeated dose, p < 0.01). However, there were no significant differences in the insulin-to-glucagon ratio and no significant change in glucose levels in these healthy subjects during the study period. Importantly, GABA significantly decreased glycated albumin levels in the repeated dosing period. Subjects with repeated dosing showed an elevated incidence of minor adverse events in comparison to placebo or the single dosing period, most notably transient discomforts such as dizziness and sore throat. However, there were no serious adverse events observed throughout the study. Our data show that GABA is rapidly absorbed and tolerated in human beings; its endocrine effects, exemplified by increasing islet hormonal secretion, suggest potential therapeutic benefits for diabetes.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Zhaoyun Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Xiaoxia Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Yi Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Fei Mao
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Junjun Mao
- Department of Pharmacy, Huashan Hospital, Fudan University Shanghai, China
| | - Xiaolan Lu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Dongdong Jiang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Jia-Ying Lv
- Department of Biostatistics, School of Public Health, Fudan University Shanghai, China
| | - Guoying Cao
- Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Institute of Antibiotics, Huashan Hospital, Fudan University Shanghai, China
| | - Jing Zhang
- Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Institute of Antibiotics, Huashan Hospital, Fudan University Shanghai, China
| | - Naiqing Zhao
- Department of Biostatistics, School of Public Health, Fudan University Shanghai, China
| | - Mark Atkinson
- Department of Pathology, College of Medicine, University of Florida Gainesville, FL, USA
| | - Dale L Greiner
- Department of Molecular Medicine, University of Massachusetts Medical School Worcester, MA, USA
| | - Gerald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, University of Toronto Toronto, ON, Canada
| | - Zheng Jiao
- Department of Pharmacy, Huashan Hospital, Fudan University Shanghai, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University Shanghai, China ; Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital Toronto, ON, Canada ; Department of Physiology and Medicine, University of Toronto ON, Canada
| |
Collapse
|
222
|
Prud'homme GJ, Glinka Y, Wang Q. Immunological GABAergic interactions and therapeutic applications in autoimmune diseases. Autoimmun Rev 2015; 14:1048-56. [DOI: 10.1016/j.autrev.2015.07.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/17/2015] [Indexed: 12/20/2022]
|
223
|
Arrojo e Drigo R, Ali Y, Diez J, Srinivasan DK, Berggren PO, Boehm BO. New insights into the architecture of the islet of Langerhans: a focused cross-species assessment. Diabetologia 2015. [PMID: 26215305 DOI: 10.1007/s00125-015-3699-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The human genome project and its search for factors underlying human diseases has fostered a major human research effort. Therefore, unsurprisingly, in recent years we have observed an increasing number of studies on human islet cells, including disease approaches focusing on type 1 and type 2 diabetes. Yet, the field of islet and diabetes research relies on the legacy of rodent-based investigations, which have proven difficult to translate to humans, particularly in type 1 diabetes. Whole islet physiology and pathology may differ between rodents and humans, and thus a comprehensive cross-species as well as species-specific view on islet research is much needed. In this review we summarise the current knowledge of interspecies islet cytoarchitecture, and discuss its potential impact on islet function and future perspectives in islet pathophysiology research.
Collapse
Affiliation(s)
- Rafael Arrojo e Drigo
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Juan Diez
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Dinesh Kumar Srinivasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital L1, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- Department of Internal Medicine 1, Ulm University Medical Centre, Ulm, Germany.
| |
Collapse
|
224
|
Jain D, Weber G, Eberhard D, Mehana AE, Eglinger J, Welters A, Bartosinska B, Jeruschke K, Weiss J, Päth G, Ariga H, Seufert J, Lammert E. DJ-1 Protects Pancreatic Beta Cells from Cytokine- and Streptozotocin-Mediated Cell Death. PLoS One 2015; 10:e0138535. [PMID: 26422139 PMCID: PMC4589499 DOI: 10.1371/journal.pone.0138535] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/01/2015] [Indexed: 01/07/2023] Open
Abstract
A hallmark feature of type 1 and type 2 diabetes mellitus is the progressive dysfunction and loss of insulin-producing pancreatic beta cells, and inflammatory cytokines are known to trigger beta cell death. Here we asked whether the anti-oxidant protein DJ-1 encoded by the Parkinson’s disease gene PARK7 protects islet cells from cytokine- and streptozotocin-mediated cell death. Wild type and DJ-1 knockout mice (KO) were treated with multiple low doses of streptozotocin (MLDS) to induce inflammatory beta cell stress and cell death. Subsequently, glucose tolerance tests were performed, and plasma insulin as well as fasting and random blood glucose concentrations were monitored. Mitochondrial morphology and number of insulin granules were quantified in beta cells. Moreover, islet cell damage was determined in vitro after streptozotocin and cytokine treatment of isolated wild type and DJ-1 KO islets using calcein AM/ethidium homodimer-1 staining and TUNEL staining. Compared to wild type mice, DJ-1 KO mice became diabetic following MLDS treatment. Insulin concentrations were substantially reduced, and fasting blood glucose concentrations were significantly higher in MLDS-treated DJ-1 KO mice compared to equally treated wild type mice. Rates of beta cell apoptosis upon MLDS treatment were twofold higher in DJ-1 KO mice compared to wild type mice, and in vitro inflammatory cytokines led to twice as much beta cell death in pancreatic islets from DJ-1 KO mice versus those of wild type mice. In conclusion, this study identified the anti-oxidant protein DJ-1 as being capable of protecting pancreatic islet cells from cell death induced by an inflammatory and cytotoxic setting.
Collapse
Affiliation(s)
- Deepak Jain
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
| | - Gesine Weber
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Amir E. Mehana
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia, Egypt
| | - Jan Eglinger
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
| | - Alena Welters
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital Düsseldorf, Düsseldorf, Germany
| | - Barbara Bartosinska
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Kay Jeruschke
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Jürgen Weiss
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
| | - Günter Päth
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
| | - Hiroyoshi Ariga
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-Ku, N12 W6, Sapporo, Japan
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
- Institute for Beta Cell Biology, German Diabetes Center at Heinrich Heine University, Leibniz Center for Diabetes Research, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf Partner Institute, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
225
|
Di Cairano ES, Moretti S, Marciani P, Sacchi VF, Castagna M, Davalli A, Folli F, Perego C. Neurotransmitters and Neuropeptides: New Players in the Control of Islet of Langerhans' Cell Mass and Function. J Cell Physiol 2015; 231:756-67. [PMID: 26332080 DOI: 10.1002/jcp.25176] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/27/2015] [Indexed: 12/19/2022]
Abstract
Islets of Langerhans control whole body glucose homeostasis, as they respond, releasing hormones, to changes in nutrient concentrations in the blood stream. The regulation of hormone secretion has been the focus of attention for a long time because it is related to many metabolic disorders, including diabetes mellitus. Endocrine cells of the islet use a sophisticate system of endocrine, paracrine and autocrine signals to synchronize their activities. These signals provide a fast and accurate control not only for hormone release but also for cell differentiation and survival, key aspects in islet physiology and pathology. Among the different categories of paracrine/autocrine signals, this review highlights the role of neurotransmitters and neuropeptides. In a manner similar to neurons, endocrine cells synthesize, accumulate, release neurotransmitters in the islet milieu, and possess receptors able to decode these signals. In this review, we provide a comprehensive description of neurotransmitter/neuropetide signaling pathways present within the islet. Then, we focus on evidence supporting the concept that neurotransmitters/neuropeptides and their receptors are interesting new targets to preserve β-cell function and mass. A greater understanding of how this network of signals works in physiological and pathological conditions would advance our knowledge of islet biology and physiology and uncover potentially new areas of pharmacological intervention. J. Cell. Physiol. 231: 756-767, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eliana S Di Cairano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Stefania Moretti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Paola Marciani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Vellea Franca Sacchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Michela Castagna
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Alberto Davalli
- Department of Internal Medicine, Diabetes and Endocrinology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas.,Department of Internal Medicine, Obesity and Comorbidities Research Center (OCRC), University of Campinas, UNICAMP, Campinas, Sao Paulo State, Brazil
| | - Carla Perego
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| |
Collapse
|
226
|
Regulation of the Neurodegenerative Process Associated to Parkinson's Disease by CD4+ T-cells. J Neuroimmune Pharmacol 2015; 10:561-75. [PMID: 26018603 DOI: 10.1007/s11481-015-9618-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/19/2015] [Indexed: 01/09/2023]
Abstract
Neuroinflammation constitutes a fundamental process involved in the physiopathology of Parkinson's disease (PD). Microglial cells play a central role in the outcome of neuroinflammation and consequent neurodegeneration of dopaminergic neurons in the substantia nigra. Current evidence indicates that CD4+ T-cells infiltrate the central nervous system (CNS) in PD, where they play a critical role determining the functional phenotype of microglia, thus regulating the progression of the neurodegenerative process. Here, we first analysed the pathogenic role of inflammatory phenotypes and the beneficial role of anti-inflammatory phenotypes of encephalitogenic CD4+ T-cells involved in the physiopathology of PD. Next, we discussed how alterations of neurotransmitter levels observed in the basal ganglia throughout the time course of PD progression could be strongly affecting the behaviour of encephalitogenic CD4+ T-cells and thereby the outcome of the neuroinflammatory process and the consequent neurodegeneration of dopaminergic neurons. Afterward, we integrated the evidence indicating the involvement of an antigen-specific immune response mediated by T-cells and B-cells against CNS-derived self-constituents in PD. Consistent with the involvement of a relevant autoimmune component in PD, we also reviewed the polymorphisms of both, class I and class II major histocompatibility complexes, associated to the risk of PD. Overall, this study gives an overview of how an autoimmune component involved in PD plays a fundamental role in the progression of the neurodegenerative process.
Collapse
|
227
|
Woraharn S, Lailerd N, Sivamaruthi BS, Wangcharoen W, Sirisattha S, Peerajan S, Chaiyasut C. Evaluation of factors that influence the L-glutamic and γ-aminobutyric acid production duringHericium erinaceusfermentation by lactic acid bacteria. CYTA - JOURNAL OF FOOD 2015. [DOI: 10.1080/19476337.2015.1042525] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
228
|
Wang P, Fiaschi-Taesch NM, Vasavada RC, Scott DK, García-Ocaña A, Stewart AF. Diabetes mellitus--advances and challenges in human β-cell proliferation. Nat Rev Endocrinol 2015; 11:201-12. [PMID: 25687999 DOI: 10.1038/nrendo.2015.9] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The treatment of diabetes mellitus represents one of the greatest medical challenges of our era. Diabetes results from a deficiency or functional impairment of insulin-producing β cells, alone or in combination with insulin resistance. It logically follows that the replacement or regeneration of β cells should reverse the progression of diabetes and, indeed, this seems to be the case in humans and rodents. This concept has prompted attempts in many laboratories to create new human β cells using stem-cell strategies to transdifferentiate or reprogramme non-β cells into β cells or to discover small molecules or other compounds that can induce proliferation of human β cells. This latter approach has shown promise, but has also proven particularly challenging to implement. In this Review, we discuss the physiology of normal human β-cell replication, the molecular mechanisms that regulate the cell cycle in human β cells, the upstream intracellular signalling pathways that connect them to cell surface receptors on β cells, the epigenetic mechanisms that control human β-cell proliferation and unbiased approaches for discovering novel molecules that can drive human β-cell proliferation. Finally, we discuss the potential and challenges of implementing strategies that replace or regenerate β cells.
Collapse
Affiliation(s)
- Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Nathalie M Fiaschi-Taesch
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
229
|
Marquard J, Otter S, Welters A, Stirban A, Fischer A, Eglinger J, Herebian D, Kletke O, Klemen MS, Stožer A, Wnendt S, Piemonti L, Köhler M, Ferrer J, Thorens B, Schliess F, Rupnik MS, Heise T, Berggren PO, Klöcker N, Meissner T, Mayatepek E, Eberhard D, Kragl M, Lammert E. Characterization of pancreatic NMDA receptors as possible drug targets for diabetes treatment. Nat Med 2015; 21:363-72. [PMID: 25774850 DOI: 10.1038/nm.3822] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/10/2015] [Indexed: 12/14/2022]
Abstract
In the nervous system, NMDA receptors (NMDARs) participate in neurotransmission and modulate the viability of neurons. In contrast, little is known about the role of NMDARs in pancreatic islets and the insulin-secreting beta cells whose functional impairment contributes to diabetes mellitus. Here we found that inhibition of NMDARs in mouse and human islets enhanced their glucose-stimulated insulin secretion (GSIS) and survival of islet cells. Further, NMDAR inhibition prolonged the amount of time that glucose-stimulated beta cells spent in a depolarized state with high cytosolic Ca(2+) concentrations. We also noticed that, in vivo, the NMDAR antagonist dextromethorphan (DXM) enhanced glucose tolerance in mice, and that in vitro dextrorphan, the main metabolite of DXM, amplified the stimulatory effect of exendin-4 on GSIS. In a mouse model of type 2 diabetes mellitus (T2DM), long-term treatment with DXM improved islet insulin content, islet cell mass and blood glucose control. Further, in a small clinical trial we found that individuals with T2DM treated with DXM showed enhanced serum insulin concentrations and glucose tolerance. Our data highlight the possibility that antagonists of NMDARs may provide a useful adjunct treatment for diabetes.
Collapse
Affiliation(s)
- Jan Marquard
- 1] Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany. [2] Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Düsseldorf, Germany
| | - Silke Otter
- 1] Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany. [2] Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany. [3] German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Alena Welters
- 1] Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany. [2] Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Düsseldorf, Germany. [3] Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany. [4] German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Alin Stirban
- Profil Institute for Metabolic Research, Neuss, Germany
| | | | - Jan Eglinger
- 1] Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany. [2] Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany. [3] German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Düsseldorf, Germany
| | - Olaf Kletke
- Institute of Neuro- and Sensory Physiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- 1] Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia. [2] Center for Open Innovations and Research, University of Maribor, Maribor, Slovenia
| | | | - Lorenzo Piemonti
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute, Milano, Italy
| | - Martin Köhler
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Jorge Ferrer
- 1] Department of Medicine, Imperial College London, London, UK. [2] Genomic Programming of Beta-Cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Marjan Slak Rupnik
- 1] Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia. [2] Center for Open Innovations and Research, University of Maribor, Maribor, Slovenia. [3] Institute of Physiology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Tim Heise
- Profil Institute for Metabolic Research, Neuss, Germany
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Nikolaj Klöcker
- Institute of Neuro- and Sensory Physiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Düsseldorf, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital Düsseldorf, Düsseldorf, Germany
| | - Daniel Eberhard
- Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Martin Kragl
- 1] Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany. [2] German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Eckhard Lammert
- 1] Institute of Metabolic Physiology, Heinrich Heine University, Düsseldorf, Germany. [2] Institute for Beta Cell Biology, German Diabetes Center, Leibniz Center for Diabetes Research, Düsseldorf, Germany. [3] German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
230
|
Auteri M, Zizzo MG, Serio R. GABA and GABA receptors in the gastrointestinal tract: from motility to inflammation. Pharmacol Res 2015; 93:11-21. [PMID: 25526825 DOI: 10.1016/j.phrs.2014.12.001] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 11/28/2014] [Accepted: 12/01/2014] [Indexed: 12/16/2022]
Abstract
Although an extensive body of literature confirmed γ-aminobutyric acid (GABA) as mediator within the enteric nervous system (ENS) controlling gastrointestinal (GI) function, the true significance of GABAergic signalling in the gut is still a matter of debate. GABAergic cells in the bowel include neuronal and endocrine-like cells, suggesting GABA as modulator of both motor and secretory GI activity. GABA effects in the GI tract depend on the activation of ionotropic GABAA and GABAC receptors and metabotropic GABAB receptors, resulting in a potential noteworthy regulation of both the excitatory and inhibitory signalling in the ENS. However, the preservation of GABAergic signalling in the gut could not be limited to the maintenance of physiologic intestinal activity. Indeed, a series of interesting studies have suggested a potential key role of GABA in the promising field of neuroimmune interaction, being involved in the modulation of immune cell activity associated with different systemic and enteric inflammatory conditions. Given the urgency of novel therapeutic strategies against chronic immunity-related pathologies, i.e. multiple sclerosis and Inflammatory Bowel Disease, an in-depth comprehension of the enteric GABAergic system in health and disease could provide the basis for new clinical application of nerve-driven immunity. Hence, in the attempt to drive novel researches addressing both the physiological and pathological importance of the GABAergic signalling in the gut, we summarized current evidence on GABA and GABA receptor function in the different parts of the GI tract, with particular focus on the potential involvement in the modulation of GI motility and inflammation.
Collapse
Affiliation(s)
- Michelangelo Auteri
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Viale delle Scienze, I-90128 Palermo, Italy
| | - Maria Grazia Zizzo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Viale delle Scienze, I-90128 Palermo, Italy
| | - Rosa Serio
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Viale delle Scienze, I-90128 Palermo, Italy.
| |
Collapse
|
231
|
Zheng J, Chen T, Zhu Y, Li HQ, Deng XL, Wang QH, Zhang JY, Chen LL. Liraglutide prevents fast weight gain and β-cell dysfunction in male catch-up growth rats. Exp Biol Med (Maywood) 2015; 240:1165-76. [PMID: 25710926 DOI: 10.1177/1535370214567614] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/03/2014] [Indexed: 12/30/2022] Open
Abstract
We reported recently that after a nutritional growth retardation, rats showed significant weight gain, central fat accumulation, dyslipidemia, and β-cell dysfunction during a catch-up growth (CUG) phase. Here, we investigated whether glucagon-like peptide-1 (GLP-1) ameliorated the rapid weight gain, central fat deposition, and β-cell dysfunction during the CUG in rats. Sixty-four male Sprague Dawley rats were stratified into four groups including normal control group, CUG group, catch-up growth with liraglutide treatment group, and catch-up growth with liraglutide and exendin 9-39 treatment group. Energy intake, body weight, and body length were monitored. Fat mass percentage was analyzed by dual energy X-ray absorptiometry scan. Plasma triglyceride and non-esterified fatty acid were measured. The β-cell mass was analyzed by morphometric analysis and signaling molecules were examined by Western blot and real-time PCR. Insulin secretion capability was evaluated by hyperglycemic clamp test. Liraglutide prevented weight gain and improved lipid and glucose metabolism in rats under CUG conditions, which were associated with reduced fasting insulin levels and improved glucose-stimulated insulin secretion. Improved β-cell function is found to be associated with increased β-cell replication as determined by β-cell density and insulin-Ki67 dual staining. Furthermore, liraglutide increased islet pancreatic duodenal homeobox-1 (Pdx-1) and B-cell lymphoma-2 transcript and protein expression, and reduced Procaspase-3 transcript and Caspase-3 p11 subunit protein expression, suggesting that expression of Pdx-1 and reduction of apoptosis may be the mechanisms involved. The therapeutic effects were attenuated in rats co-administered with exendin 9-39, suggesting a GLP-1 receptor-dependent mechanism. These studies revealed that incretin therapy effectively prevented fast weight gain and β-cell dysfunction in rats under conditions of nutrition restriction followed by nutrition excess, which is in part due to enhanced functional β-cell mass and insulin secretory capacity.
Collapse
Affiliation(s)
- Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M2J4A6, Canada
| | - Ting Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China Department of Endocrinology, Soochow University Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215000, China
| | - Ying Zhu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hui-Qing Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xiu-Ling Deng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Qing-Hua Wang
- Division of Endocrinology and Metabolism, The Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario M2J4A6, Canada Departments of Physiology and Medicine, University of Toronto, Toronto M2J4A6, Canada
| | - Jiao-Yue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lu-Lu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
232
|
Chandravanshi B, Dhanushkodi A, Bhonde R. High Recovery of Functional Islets Stored at Low and Ultralow Temperatures. Rev Diabet Stud 2015; 11:267-78. [PMID: 26177487 DOI: 10.1900/rds.2014.11.267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Poor recovery of islets upon cryopreservation is the main hurdle in islet banking. Pancreatic islets have a poor antioxidative defense mechanism, and exposure of islets to low temperature leads to oxidative stress. AIM We aimed to investigate whether known compounds such as metformin, γ aminobutyric acid (GABA), docosahexanoic acid (DHA), or eicosapentaenoic acid (EPA) alone or in combination are capable of reducing oxidative stress for better islet recovery upon storage at suboptimal temperatures. METHODS Islets isolated from mouse pancreas were stored at low temperature (4°C) for 15 days and at ultralow temperature (-196°C) for 30 days with or without additives. After revival from cold storage, islets were assessed by using three methods: (1) specificity by dithizone (DTZ), (2) viability by fluorescein diacetate/propidium iodide (FDA/PI) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra-zolium bromide (MTT) assay, and (3) functionality by glucose-stimulated insulin secretion (GSIS). The oxidative status of the islets stored at suboptimal temperatures was determined by both intracellular free radical release (fluorometric analysis) and lipid peroxidation (enzymatic determination). RESULTS Supplementation with additives led to an improvement in islet survival upon storage at suboptimal temperatures, without depletion of insulin secretory activity, which was comparable to that of controls. The additives acted as cryoprotectants and antioxidants as revealed by high recovery of viable islets and reduction in total reactive oxygen species (ROS) and malonidealdehyde (MDA), respectively. CONCLUSIONS Our results demonstrate for the first time that supplementation with EPA, DHA, and metformin may lead to higher islet recovery from -196°C storage, enabling proper islet banking.
Collapse
Affiliation(s)
- Bhawna Chandravanshi
- School of Regenerative Medicine, Manipal University, Bellary Road, Yelahanka, Bangalore 560065, India
| | - Anandh Dhanushkodi
- School of Regenerative Medicine, Manipal University, Bellary Road, Yelahanka, Bangalore 560065, India
| | - Ramesh Bhonde
- School of Regenerative Medicine, Manipal University, Bellary Road, Yelahanka, Bangalore 560065, India
| |
Collapse
|
233
|
Kanaani J, Cianciaruso C, Phelps EA, Pasquier M, Brioudes E, Billestrup N, Baekkeskov S. Compartmentalization of GABA synthesis by GAD67 differs between pancreatic beta cells and neurons. PLoS One 2015; 10:e0117130. [PMID: 25647668 PMCID: PMC4315522 DOI: 10.1371/journal.pone.0117130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/19/2014] [Indexed: 11/22/2022] Open
Abstract
The inhibitory neurotransmitter GABA is synthesized by the enzyme glutamic acid decarboxylase (GAD) in neurons and in pancreatic β-cells in islets of Langerhans where it functions as a paracrine and autocrine signaling molecule regulating the function of islet endocrine cells. The localization of the two non-allelic isoforms GAD65 and GAD67 to vesicular membranes is important for rapid delivery and accumulation of GABA for regulated secretion. While the membrane anchoring and trafficking of GAD65 are mediated by intrinsic hydrophobic modifications, GAD67 remains hydrophilic, and yet is targeted to vesicular membrane pathways and synaptic clusters in neurons by both a GAD65-dependent and a distinct GAD65-independent mechanism. Herein we have investigated the membrane association and targeting of GAD67 and GAD65 in monolayer cultures of primary rat, human, and mouse islets and in insulinoma cells. GAD65 is primarily detected in Golgi membranes and in peripheral vesicles distinct from insulin vesicles in β-cells. In the absence of GAD65, GAD67 is in contrast primarily cytosolic in β-cells; its co-expression with GAD65 is necessary for targeting to Golgi membranes and vesicular compartments. Thus, the GAD65-independent mechanism for targeting of GAD67 to synaptic vesicles in neurons is not functional in islet β-cells. Therefore, only GAD65:GAD65 homodimers and GAD67:GAD65 heterodimers, but not the GAD67:GAD67 homodimer gain access to vesicular compartments in β-cells to facilitate rapid accumulation of newly synthesized GABA for regulated secretion and fine tuning of GABA-signaling in islets of Langerhans.
Collapse
Affiliation(s)
- Jamil Kanaani
- Departments of Medicine and Microbiology/Immunology, Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Chiara Cianciaruso
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Edward A. Phelps
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Miriella Pasquier
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Estelle Brioudes
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Nils Billestrup
- Institute of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steinunn Baekkeskov
- Departments of Medicine and Microbiology/Immunology, Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
234
|
Li Q, Lai ZC. Recent progress in studies of factors that elicit pancreatic β-cell expansion. Protein Cell 2015; 6:81-7. [PMID: 25492376 PMCID: PMC4312764 DOI: 10.1007/s13238-014-0123-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 11/13/2014] [Indexed: 12/26/2022] Open
Abstract
The loss of or decreased functional pancreatic β-cell is a major cause of type 1 and type 2 diabetes. Previous studies have shown that adult β-cells can maintain their ability for a low level of turnover through replication and neogenesis. Thus, a strategy to prevent and treat diabetes would be to enhance the ability of β-cells to increase the mass of functional β-cells. Consequently, much effort has been devoted to identify factors that can effectively induce β-cell expansion. This review focuses on recent reports on small molecules and protein factors that have been shown to promote β-cell expansion.
Collapse
Affiliation(s)
- Qiu Li
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021 China
| | - Zhi-Chun Lai
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021 China
- Department of Biology, The Pennsylvania State University, University Park, PA 16802 USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
235
|
Wan Y, Wang Q, Prud’homme GJ. GABAergic system in the endocrine pancreas: a new target for diabetes treatment. Diabetes Metab Syndr Obes 2015; 8:79-87. [PMID: 25678807 PMCID: PMC4322886 DOI: 10.2147/dmso.s50642] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Excessive loss of functional pancreatic β-cell mass, mainly due to apoptosis, is a major factor in the development of hyperglycemia in both type 1 and type 2 diabetes (T1D and T2D). In T1D, β-cells are destroyed by immunological mechanisms. In T2D, while metabolic factors are known to contribute to β-cell failure and subsequent apoptosis, mounting evidence suggests that islet inflammation also plays an important role in the loss of β-cell mass. Therefore, it is of great importance for clinical intervention to develop new therapies. γ-Aminobutyric acid (GABA), a major neurotransmitter, is also produced by islet β-cells, where it functions as an important intraislet transmitter in regulating islet-cell secretion and function. Importantly, recent studies performed in rodents, including in vivo studies of xenotransplanted human islets, reveal that GABA exerts β-cell regenerative effects. Moreover, it protects β-cells against apoptosis induced by cytokines, drugs, and other stresses, and has anti-inflammatory and immunoregulatory activities. It ameliorates the manifestations of diabetes in preclinical models, suggesting potential applications for the treatment of diabetic patients. This review outlines the actions of GABA relevant to β-cell regeneration, including its signaling mechanisms and potential interactions with other mediators. These studies increase our understanding of the regenerative processes of pancreatic β-cells, and help pave the way for the development of regenerative medicine for diabetes.
Collapse
Affiliation(s)
- Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Medical College, Fudan University, Shanghai, People’s Republic of China
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St Michael’s Hospital, Toronto, ON, Canada
- Departments of Physiology and Medicine, Faculty of Medicine, Toronto, ON, Canada
- Correspondence: Qinghua Wang, Division of Endocrinology and Metabolism, St Michael’s Hospital, 30 Bond Street, Toronto, ON M5B 1W8, Canada, Tel +1 416 864 6060 ext 77 610, Fax +1 416 864 5140, Email
| | - Gerald J Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
236
|
Wang Y, Liu M, Zhao L, Qiu Y, Zhuang Y. Interactions of γ-aminobutyric acid and whey proteins/caseins during fortified milk production. RSC Adv 2015. [DOI: 10.1039/c5ra16271h] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
γ-aminobutyric acid mainly cross-links the β-lg fraction and adducts with α-la or αs1-casein fractions, and tends to form its linear or membered ring structure oligomers.
Collapse
Affiliation(s)
- Yaosong Wang
- State Key Laboratory of Bioreactor Engineering
- R&D Center of Separation and Extraction Technology in Fermentation Industry
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Mingying Liu
- State Key Laboratory of Bioreactor Engineering
- R&D Center of Separation and Extraction Technology in Fermentation Industry
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Liming Zhao
- State Key Laboratory of Bioreactor Engineering
- R&D Center of Separation and Extraction Technology in Fermentation Industry
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Yongjun Qiu
- State Key Laboratory of Bioreactor Engineering
- R&D Center of Separation and Extraction Technology in Fermentation Industry
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering
- R&D Center of Separation and Extraction Technology in Fermentation Industry
- East China University of Science and Technology
- Shanghai 200237
- China
| |
Collapse
|
237
|
Kook MC, Cho SC, Kang J, Song Y, Park H. Effect of gamma-aminobutyric acid produced by Lactobacillus sakei B2-16 on diet and exercise in high fat diet-induced Obese rats. Food Sci Biotechnol 2014. [DOI: 10.1007/s10068-014-0268-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
238
|
Purwana I, Zheng J, Li X, Deurloo M, Son DO, Zhang Z, Liang C, Shen E, Tadkase A, Feng ZP, Li Y, Hasilo C, Paraskevas S, Bortell R, Greiner DL, Atkinson M, Prud'homme GJ, Wang Q. GABA promotes human β-cell proliferation and modulates glucose homeostasis. Diabetes 2014; 63:4197-205. [PMID: 25008178 DOI: 10.2337/db14-0153] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
γ-Aminobutyric acid (GABA) exerts protective and regenerative effects on mouse islet β-cells. However, in humans it is unknown whether it can increase β-cell mass and improve glucose homeostasis. To address this question, we transplanted a suboptimal mass of human islets into immunodeficient NOD-scid-γ mice with streptozotocin-induced diabetes. GABA treatment increased grafted β-cell proliferation, while decreasing apoptosis, leading to enhanced β-cell mass. This was associated with increased circulating human insulin and reduced glucagon levels. Importantly, GABA administration lowered blood glucose levels and improved glucose excursion rates. We investigated GABA receptor expression and signaling mechanisms. In human islets, GABA activated a calcium-dependent signaling pathway through both GABA A receptor and GABA B receptor. This activated the phosphatidylinositol 3-kinase-Akt and CREB-IRS-2 signaling pathways that convey GABA signals responsible for β-cell proliferation and survival. Our findings suggest that GABA regulates human β-cell mass and may be beneficial for the treatment of diabetes or improvement of islet transplantation.
Collapse
Affiliation(s)
- Indri Purwana
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Juan Zheng
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xiaoming Li
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marielle Deurloo
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Dong Ok Son
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhaoyun Zhang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Christie Liang
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eddie Shen
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Akshaya Tadkase
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Zhong-Ping Feng
- Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yiming Li
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Craig Hasilo
- Department of Surgery, McGill University, and Human Islet Transplantation Laboratory, McGill University Health Centre, Montreal, Quebec, Canada
| | - Steven Paraskevas
- Department of Surgery, McGill University, and Human Islet Transplantation Laboratory, McGill University Health Centre, Montreal, Quebec, Canada
| | - Rita Bortell
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Dale L Greiner
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Health Science Center, Gainesville, FL
| | - Gerald J Prud'homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Qinghua Wang
- Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada Departments of Physiology and Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
239
|
Yang SN, Shi Y, Yang G, Li Y, Yu J, Berggren PO. Ionic mechanisms in pancreatic β cell signaling. Cell Mol Life Sci 2014; 71:4149-77. [PMID: 25052376 PMCID: PMC11113777 DOI: 10.1007/s00018-014-1680-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 07/03/2014] [Accepted: 07/10/2014] [Indexed: 01/07/2023]
Abstract
The function and survival of pancreatic β cells critically rely on complex electrical signaling systems composed of a series of ionic events, namely fluxes of K(+), Na(+), Ca(2+) and Cl(-) across the β cell membranes. These electrical signaling systems not only sense events occurring in the extracellular space and intracellular milieu of pancreatic islet cells, but also control different β cell activities, most notably glucose-stimulated insulin secretion. Three major ion fluxes including K(+) efflux through ATP-sensitive K(+) (KATP) channels, the voltage-gated Ca(2+) (CaV) channel-mediated Ca(2+) influx and K(+) efflux through voltage-gated K(+) (KV) channels operate in the β cell. These ion fluxes set the resting membrane potential and the shape, rate and pattern of firing of action potentials under different metabolic conditions. The KATP channel-mediated K(+) efflux determines the resting membrane potential and keeps the excitability of the β cell at low levels. Ca(2+) influx through CaV1 channels, a major type of β cell CaV channels, causes the upstroke or depolarization phase of the action potential and regulates a wide range of β cell functions including the most elementary β cell function, insulin secretion. K(+) efflux mediated by KV2.1 delayed rectifier K(+) channels, a predominant form of β cell KV channels, brings about the downstroke or repolarization phase of the action potential, which acts as a brake for insulin secretion owing to shutting down the CaV channel-mediated Ca(2+) entry. These three ion channel-mediated ion fluxes are the most important ionic events in β cell signaling. This review concisely discusses various ionic mechanisms in β cell signaling and highlights KATP channel-, CaV1 channel- and KV2.1 channel-mediated ion fluxes.
Collapse
Affiliation(s)
- Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-171 76, Stockholm, Sweden,
| | | | | | | | | | | |
Collapse
|
240
|
Kitagishi Y, Nakanishi A, Minami A, Asai Y, Yasui M, Iwaizako A, Suzuki M, Ono Y, Ogura Y, Matsuda S. Certain Diet and Lifestyle May Contribute to Islet β-cells Protection in Type-2 Diabetes via the Modulation of Cellular PI3K/AKT Pathway. Open Biochem J 2014; 8:74-82. [PMID: 25400709 PMCID: PMC4231374 DOI: 10.2174/1874091x01408010074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/13/2014] [Accepted: 08/18/2014] [Indexed: 12/18/2022] Open
Abstract
PI3K/AKT pathway has been shown to play a pivotal role on islet β-cell protection, enhancing β-cell survival by stimulating cell proliferation and inhibiting cell apoptosis. Accordingly, this pathway appears to be crucial in type-2 diabetes. Understanding the regulations of this pathway may provide a better efficacy of new therapeutic approaches. In this review, we summarize advances on the involvement of the PI3K/AKT pathway in hypothetical intra-cellular signaling of islet β-cells. As recent findings may show the nutritional regulation of the survival pathway in the islet β-cells through activation of the PI3K/AKT pathway, we also review studies on the features of several diets, correlated lifestyle, and its signaling pathway involved in type-2 diabetes. The molecular mechanisms contributing to the disease are the subject of considerable investigation, as a better understanding of the pathogenesis will lead to novel therapies against a condition of the disease.
Collapse
Affiliation(s)
- Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Atsuko Nakanishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Akari Minami
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yurina Asai
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Mai Yasui
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Akiko Iwaizako
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Miho Suzuki
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yuna Ono
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yasunori Ogura
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
241
|
Wang S, Luo Y, Feng A, Li T, Yang X, Nofech-Mozes R, Yu M, Wang C, Li Z, Yi F, Liu C, Lu WY. Ethanol induced impairment of glucose metabolism involves alterations of GABAergic signaling in pancreatic β-cells. Toxicology 2014; 326:44-52. [PMID: 25456265 DOI: 10.1016/j.tox.2014.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/09/2014] [Accepted: 10/10/2014] [Indexed: 01/02/2023]
Abstract
Alcohol overindulgence is a risk factor of type 2 diabetes mellitus. However, the mechanisms by which alcohol overindulgence damages glucose metabolism remain unclear. Pancreatic islet β-cells are endowed with type-A γ-aminobutyric acid receptor (GABAAR) mediated autocrine signaling mechanism, which regulates insulin secretion and fine-tunes glucose metabolism. In neurons GABAAR is one of the major targets for alcohol. This study investigated whether ethanol alters glucose metabolism by affecting GABAAR signaling in pancreatic β-cells. Blood glucose level of test mice was measured using a blood glucose meter. Insulin secretion by the pancreatic β-cell line INS-1 cells was examined using a specific insulin ELISA kit. Whole-cell patch-clamp recording was used to evaluate GABA-elicited current in INS-1 cells. Western blot and immunostaining were used to measure the expression of GABAAR subunits in mouse pancreatic tissues or in INS-1 cells. Intraperitoneal (i.p.) administration of ethanol (3.0g/kg body weight) to mice altered glucose metabolism, which was associated with decreased expression of GABAAR α1- and δ- subunits on the surface of pancreatic β-cells. Acute treatment of cultured INS-1cells with ethanol (60mM) decreased the GABA-induced current and reduced insulin secretion. In contrast, treating INS-1 cells with GABA (100μM) largely prevented the ethanol-induced reduction of insulin release. Importantly, pre-treating mice with GABA (i.p., 1.5mg/kg body weight) partially reversed ethanol-induced impairment of glucose homeostasis in mice. Our data suggest a novel role of pancreatic GABA signaling in protecting pancreatic islet β-cells from ethanol-induced dysfunction.
Collapse
Affiliation(s)
- Shuanglian Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Yan Luo
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Allen Feng
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Tao Li
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Xupeng Yang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Roy Nofech-Mozes
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada
| | - Meng Yu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Changhui Wang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Ziwei Li
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Chuanyong Liu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, PR China.
| | - Wei-Yang Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada; Robarts Research Institute, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
242
|
Williams DB. Inhibitory effects of insulin on GABAAcurrents modulated by the GABAAalpha subunit. J Recept Signal Transduct Res 2014; 35:516-22. [DOI: 10.3109/10799893.2014.960935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
243
|
GABA protects pancreatic beta cells against apoptosis by increasing SIRT1 expression and activity. Biochem Biophys Res Commun 2014; 452:649-54. [PMID: 25193706 DOI: 10.1016/j.bbrc.2014.08.135] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 08/25/2014] [Indexed: 02/08/2023]
Abstract
We have previously shown that GABA protects pancreatic islet cells against apoptosis and exerts anti-inflammatory effects. Notably, GABA inhibited the activation of NF-κB in both islet cells and lymphocytes. NF-κB activation is detrimental to beta cells by promoting apoptosis. However, the mechanisms by which GABA mediates these effects are unknown. Because the above-mentioned effects mimic the activity of sirtuin 1 (SIRT1) in beta cells, we investigated whether it is involved. SIRT1 is an NAD(+)-dependent deacetylase that enhances insulin secretion, and counteracts inflammatory signals in beta cells. We found that the incubation of a clonal beta-cell line (rat INS-1) with GABA increased the expression of SIRT1, as did GABA receptor agonists acting on either type A or B receptors. NAD(+) (an essential cofactor of SIRT1) was also increased. GABA augmented SIRT1 enzymatic activity, which resulted in deacetylation of the p65 component of NF-κB, and this is known to interfere with the activation this pathway. GABA increased insulin production and reduced drug-induced apoptosis, and these actions were reversed by SIRT1 inhibitors. We examined whether SIRT1 is similarly induced in newly isolated human islet cells. Indeed, GABA increased both NAD(+) and SIRT1 (but not sirtuins 2, 3 and 6). It protected human islet cells against spontaneous apoptosis in culture, and this was negated by a SIRT1 inhibitor. Thus, our findings suggest that major beneficial effects of GABA on beta cells are due to increased SIRT1 and NAD(+), and point to a new pathway for diabetes therapy.
Collapse
|
244
|
Tian J, Dang H, Nguyen AV, Chen Z, Kaufman DL. Combined therapy with GABA and proinsulin/alum acts synergistically to restore long-term normoglycemia by modulating T-cell autoimmunity and promoting β-cell replication in newly diabetic NOD mice. Diabetes 2014; 63:3128-34. [PMID: 25146474 PMCID: PMC4141368 DOI: 10.2337/db13-1385] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Antigen-based therapies (ABTs) fail to restore normoglycemia in newly diabetic NOD mice, perhaps because too few β-cells remain by the time that ABT-induced regulatory responses arise and spread. We hypothesized that combining a fast-acting anti-inflammatory agent with an ABT could limit pathogenic responses while ABT-induced regulatory responses arose and spread. γ-Aminobutyric acid (GABA) administration can inhibit inflammation, enhance regulatory T-cell (Treg) responses, and promote β-cell replication in mice. We examined the effect of combining a prototypic ABT, proinsulin/alum, with GABA treatment in newly diabetic NOD mice. Proinsulin/alum monotherapy failed to correct hyperglycemia, while GABA monotherapy restored normoglycemia for a short period. Combined treatment restored normoglycemia in the long term with apparent permanent remission in some mice. Proinsulin/alum monotherapy induced interleukin (IL)-4- and IL-10-secreting T-cell responses that spread to other β-cell autoantigens. GABA monotherapy induced moderate IL-10 (but not IL-4) responses to β-cell autoantigens. Combined treatment synergistically reduced spontaneous type 1 T-helper cell responses to autoantigens, ABT-induced IL-4 and humoral responses, and insulitis, but enhanced IL-10 and Treg responses and promoted β-cell replication in the islets. Thus, combining ABT with GABA can inhibit pathogenic T-cell responses, induce Treg responses, promote β-cell replication, and effectively restore normoglycemia in newly diabetic NOD mice. Since these treatments appear safe for humans, they hold promise for type 1 diabetes intervention.
Collapse
Affiliation(s)
- Jide Tian
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - Hoa Dang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - An Viet Nguyen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - Zheying Chen
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| | - Daniel L Kaufman
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
245
|
Zhang X, Du Z, Liu J, He J. Γ-aminobutyric acid receptors affect the progression and migration of tumor cells. J Recept Signal Transduct Res 2014; 34:431-9. [DOI: 10.3109/10799893.2013.856918] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
246
|
Paul AM, Branton WG, Walsh JG, Polyak MJ, Lu JQ, Baker GB, Power C. GABA transport and neuroinflammation are coupled in multiple sclerosis: regulation of the GABA transporter-2 by ganaxolone. Neuroscience 2014; 273:24-38. [PMID: 24814730 DOI: 10.1016/j.neuroscience.2014.04.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 04/15/2014] [Accepted: 04/17/2014] [Indexed: 10/25/2022]
Abstract
Interactions between neurotransmitters and the immune system represent new prospects for understanding neuroinflammation and associated neurological disease. GABA is the chief inhibitory neurotransmitter but its actions on immune pathways in the brain are unclear. In the present study, we investigated GABAergic transport in conjunction with neuroinflammation in models of multiple sclerosis (MS). Protein and mRNA levels of γ-amino butyric acid transporter 2 (GAT-2) were examined in cerebral white matter from MS and control (Non-MS) patients, in cultured human macrophages, microglia and astrocytes, and in spinal cords from mice with and without experimental autoimmune encephalomyelitis (EAE) using western blotting, immunocytochemistry and quantitative real-time polymerase chain reaction (qRT-PCR). GABA levels were measured by HPLC. The GAT-2's expression was increased in MS patients' (n=6) white matter, particularly in macrophage lineage cells, compared to Non-MS patients (n=6) (p<0.05). Interferon-γ (IFN-γ) stimulation of human macrophage lineage cells induced GAT-2 expression and reduced extracellular GABA levels (p<0.05) but soluble GABA treatment suppressed HLA-DRα, GAT-2 and XBP-1/s expression in stimulated macrophage lineage cells (p<0.05). Similarly, the synthetic allopregnanolone analog, ganaxolone (GNX), repressed GAT-2, JAK-1 and STAT-1 expression in activated macrophage lineage cells (p<0.05). In vivo GNX treatment reduced Gat-2, Cd3ε, MhcII, and Xbp-1/s expression in spinal cords following EAE induction (p<0.05), which was correlated with improved neurobehavioral outcomes and reduced neuroinflammation, demyelination and axonal injury. These findings highlight altered GABAergic transport through GAT-2 induction during neuroinflammation. GABA transport and neuroinflammation are closely coupled but regulated by GNX, pointing to GABAergic pathways as therapeutic targets in neuroinflammatory diseases.
Collapse
Affiliation(s)
- A M Paul
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - W G Branton
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - J G Walsh
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - M J Polyak
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - J-Q Lu
- Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada
| | - G B Baker
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - C Power
- Department of Medicine, University of Alberta, Edmonton, AB, Canada; Department of Laboratory Medicine & Pathology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
247
|
Oda K, Imanishi T, Yamane Y, Ueno Y, Mori Y. Bio-functional pickles that reduce blood pressure of rats. Biosci Biotechnol Biochem 2014; 78:882-90. [DOI: 10.1080/09168451.2014.893187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Abstract
Addition of γ-aminobutyric acid (GABA) and angiotensin converting enzyme (ACE)-inhibitory peptides to the pickles was studied in order to develop a new type of pickles that reduce blood pressure. Based on the outcome of these studies, a new type of fermentation bed composed of rice bran and white miso has been successfully developed. The advantage of such pickles is that they not only contain both GABA and ACE-inhibitory peptides, but also that their taste and flavor are excellent, with colors close to the original ones. The new type of pickles could temporarily reduce blood pressure in two types of rats, spontaneously hypertensive rats and NaCl-sensitive model rats. Thus, the newly developed pickles appear to be beneficial for pickle business.
Collapse
Affiliation(s)
- Kohei Oda
- Kyoto Institute of Technology, Sakyo-ku, Kyoto, Japan
| | | | | | - Yoshie Ueno
- Kyoto Prefectural Technology Center for Small and Medium Enterprises, Shimogyo-ku, Kyoto, Japan
| | | |
Collapse
|
248
|
Gamma-aminobutyric acid improves oxidative stress and function of the thyroid in high-fat diet fed mice. J Funct Foods 2014. [DOI: 10.1016/j.jff.2014.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
249
|
Li M, Song LJ, Qin XY. Advances in the cellular immunological pathogenesis of type 1 diabetes. J Cell Mol Med 2014; 18:749-58. [PMID: 24629100 PMCID: PMC4119381 DOI: 10.1111/jcmm.12270] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/30/2014] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is an autoimmune disease caused by the immune-mediated destruction of insulin-producing pancreatic β cells. In recent years, the incidence of type 1 diabetes continues to increase. It is supposed that genetic, environmental and immune factors participate in the damage of pancreatic β cells. Both the immune regulation and the immune response are involved in the pathogenesis of type 1 diabetes, in which cellular immunity plays a significant role. For the infiltration of CD4(+) and CD8(+) T lymphocyte, B lymphocytes, natural killer cells, dendritic cells and other immune cells take part in the damage of pancreatic β cells, which ultimately lead to type 1 diabetes. This review outlines the cellular immunological mechanism of type 1 diabetes, with a particular emphasis to T lymphocyte and natural killer cells, and provides the effective immune therapy in T1D, which is approached at three stages. However, future studies will be directed at searching for an effective, safe and long-lasting strategy to enhance the regulation of a diabetogenic immune system with limited toxicity and without global immunosuppression.
Collapse
Affiliation(s)
- Min Li
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Lu-Jun Song
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| | - Xin-Yu Qin
- Department of General Surgery, Zhongshan Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
250
|
Ardestani A, Paroni F, Azizi Z, Kaur S, Khobragade V, Yuan T, Frogne T, Tao W, Oberholzer J, Pattou F, Conte JK, Maedler K. MST1 is a key regulator of beta cell apoptosis and dysfunction in diabetes. Nat Med 2014; 20:385-397. [PMID: 24633305 PMCID: PMC3981675 DOI: 10.1038/nm.3482] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/21/2014] [Indexed: 12/31/2022]
Abstract
Apoptotic cell death is a hallmark of the loss of insulin producing beta-cells in all forms of diabetes mellitus. Current treatment fails to halt the decline in functional beta-cell mass. Strategies to prevent beta-cell apoptosis and dysfunction are urgently needed. Here, we identified Mammalian Sterile 20-like kinase 1 (MST1) as a critical regulator of apoptotic beta-cell death and function. MST1 was strongly activated in beta-cells under diabetogenic conditions and correlated with beta-cell apoptosis. MST1 specifically induced the mitochondrial-dependent pathway of apoptosis in beta-cells through up-regulation of the BH3-only protein Bim. MST1 directly phosphorylated PDX1 at Thr11, resulting in its ubiquitination, degradation and impaired insulin secretion. Mst1 deficiency completely restored normoglycemia, beta-cell function and survival in vitro and in vivo. We show MST1 as novel pro-apoptotic kinase and key mediator of apoptotic signaling and beta-cell dysfunction, which may serve as target for the development of novel therapies for diabetes.
Collapse
Affiliation(s)
- Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, Germany
| | - Federico Paroni
- Centre for Biomolecular Interactions Bremen, University of Bremen, Germany
| | - Zahra Azizi
- Centre for Biomolecular Interactions Bremen, University of Bremen, Germany
| | - Supreet Kaur
- Centre for Biomolecular Interactions Bremen, University of Bremen, Germany
| | | | - Ting Yuan
- Centre for Biomolecular Interactions Bremen, University of Bremen, Germany
| | - Thomas Frogne
- Department of Beta-cell Regeneration, Hagedorn Research Institute, Gentofte, Denmark
| | - Wufan Tao
- Institute of Developmental Biology and Molecular Medicine, Fudan University, Shanghai, China
| | - Jose Oberholzer
- Division of Transplantation, University of Illinois at Chicago, IL, USA
| | - Francois Pattou
- Thérapie Cellulaire du Diabète, INSERM /Université de Lille Nord de France, France
| | - Julie Kerr Conte
- Thérapie Cellulaire du Diabète, INSERM /Université de Lille Nord de France, France
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, Germany
| |
Collapse
|