1
|
Primavera R, Wang J, Buchwald P, Ganguly A, Patel S, Bettencourt L, Chetty S, Yarani R, Regmi S, Levitte S, Kevadiya B, Guindani M, Decuzzi P, Thakor AS. Controlled Nutrient Delivery to Pancreatic Islets Using Polydopamine-Coated Mesoporous Silica Nanoparticles. NANO LETTERS 2025; 25:939-950. [PMID: 39791700 DOI: 10.1021/acs.nanolett.4c03613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In this study, we designed a nanoscale platform for sustained amino acid delivery to support transplanted pancreatic islets. The platform features mesoporous silica nanoparticles (MSNPs) loaded with glutamine (G), an essential amino acid required for islet survival and function, and coated with polydopamine (PD). We investigated various PD concentrations (0.5-2 mg/mL) and incubation times (0.5-2 h) to optimize G release, identifying that a PD concentration of 0.5 mg/mL incubated for 0.5 h yielded the best results to support islet viability and functionality ex vivo, particularly under inflammatory conditions. In syngeneic islet transplantation in STZ-diabetic mice, G alone provided only temporary benefits; however, PD-G-MSNPs significantly improved islet engraftment and function, with animals maintaining glycemic control for 30 days due to controlled G release. Our findings support the use of this nanoscale platform to provide essential nutrients like G to transplanted islets until they can establish their own blood and nutrient supply.
Collapse
Affiliation(s)
- Rosita Primavera
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Jing Wang
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Abantika Ganguly
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shaini Patel
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Lili Bettencourt
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Shashank Chetty
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Shobha Regmi
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Steven Levitte
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Bhavesh Kevadiya
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Michele Guindani
- Department of Biostatistics, Jonathan and Karin Fielding School of Public Health, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, Genoa 16163, Italy
| | - Avnesh S Thakor
- Department of Radiology, Interventional Radiology Innovation at Stanford (IRIS), Stanford University School of Medicine, Palo Alto, California 94304, United States
| |
Collapse
|
2
|
Lu J, Zhao RX, Xiong FR, Zhu JJ, Shi TT, Zhang YC, Peng GX, Yang JK. All-potassium channel CRISPR screening reveals a lysine-specific pathway of insulin secretion. Mol Metab 2024; 80:101885. [PMID: 38246588 PMCID: PMC10847698 DOI: 10.1016/j.molmet.2024.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
OBJECTIVE Genome-scale CRISPR-Cas9 knockout coupled with single-cell RNA sequencing (scRNA-seq) has been used to identify function-related genes. However, this method may knock out too many genes, leading to low efficiency in finding genes of interest. Insulin secretion is controlled by several electrophysiological events, including fluxes of KATP depolarization and K+ repolarization. It is well known that glucose stimulates insulin secretion from pancreatic β-cells, mainly via the KATP depolarization channel, but whether other nutrients directly regulate the repolarization K+ channel to promote insulin secretion is unknown. METHODS We used a system involving CRISPR-Cas9-mediated knockout of all 83 K+ channels and scRNA-seq in a pancreatic β cell line to identify genes associated with insulin secretion. RESULTS The expression levels of insulin genes were significantly increased after all-K+ channel knockout. Furthermore, Kcnb1 and Kcnh6 were the two most important repolarization K+ channels for the increase in high-glucose-dependent insulin secretion that occurred upon application of specific inhibitors of the channels. Kcnh6 currents, but not Kcnb1 currents, were reduced by one of the amino acids, lysine, in both transfected cells, primary cells and mice with β-cell-specific deletion of Kcnh6. CONCLUSIONS Our function-related CRISPR screen with scRNA-seq identifies Kcnh6 as a lysine-specific channel.
Collapse
Affiliation(s)
- Jing Lu
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Ru-Xuan Zhao
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Feng-Ran Xiong
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Juan-Juan Zhu
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Ting-Ting Shi
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Ying-Chao Zhang
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China
| | - Gong-Xin Peng
- Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing 100740, China
| | - Jin-Kui Yang
- Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Diabetes Research and Care, Beijing 100730, China.
| |
Collapse
|
3
|
Liu H, Niu T, Qiu G, Cui S, Zhang D. Taurine promotes insulin synthesis by enhancing Isl-1 expression through miR-7a/RAF1/ERK1/2 pathway. In Vitro Cell Dev Biol Anim 2024; 60:23-35. [PMID: 38117455 DOI: 10.1007/s11626-023-00835-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/04/2023] [Indexed: 12/21/2023]
Abstract
It has been well established that the circulating taurine affects the insulin synthesis in pancreatic islet β-cells, whereas miR-7a and LIM-homeodomain transcription factor Isl-1 are important intracellular factors regulating insulin transcription and synthesis. However, it still remains unknown whether taurine regulates insulin synthesis by affecting miR-7a and/or Isl-1 expressions in mouse pancreatic islet β-cells. The present study was thus proposed to identify the effects of taurine on the expressions of miR-7a and/or Isl-1 and their relations to insulin synthesis in mouse pancreatic islet β-cells by using miR-7a2 knockout (KO) and taurine transporter (TauT) KO mouse models and the related in vitro experiments. The results demonstrated that taurine supplement significantly decreased the pancreas miR-7a expression, but sharply upregulated the pancreas Isl-1 and insulin expressions, and serum insulin levels. However, the enhanced effects of taurine on Isl-1 expression and insulin synthesis were mitigated in the TauT KO and miR-7a2 KO mice. In addition, our results confirmed that taurine markedly increased pancreas RAF1 and ERK1/2 expressions. Collectively, the present study firstly demonstrates that taurine regulates insulin synthesis through TauT/miR-7a/RAF1/ERK1/2/Isl-1 signaling pathway, which are crucial for our understanding the mechanisms of taurine affecting insulin synthesis, and also potential for establishing the therapeutic strategies for diabetes and the diseases related to metabolism.
Collapse
Affiliation(s)
- Hui Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Tongjuan Niu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Guobin Qiu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China
| | - Di Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, Jiangsu, People's Republic of China.
| |
Collapse
|
4
|
Lee C, Lee DK, Wei IA, Qiu TA, Rubakhin SS, Roper MG, Sweedler JV. Relations between Glucose and d-Amino Acids in the Modulation of Biochemical and Functional Properties of Rodent Islets of Langerhans. ACS OMEGA 2023; 8:47723-47734. [PMID: 38144114 PMCID: PMC10733910 DOI: 10.1021/acsomega.3c05983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 12/26/2023]
Abstract
The cell-to-cell signaling role of d-amino acids (d-AAs) in the mammalian endocrine system, particularly in the islets of Langerhans, has drawn growing interest for their potential involvement in modulating glucose metabolism. Previous studies found colocalization of serine racemase [produces d-serine (d-Ser)] and d-alanine (d-Ala) within insulin-secreting beta cells and d-aspartate (d-Asp) within glucagon-secreting alpha cells. Expressed in the islets, functional N-methyl-d-aspartate receptors are involved in the modulation of glucose-stimulated insulin secretion and have binding sites for several d-AAs. However, knowledge of the regulation of d-AA levels in the islets during glucose stimulation as well as the response of islets to different levels of extracellular d-AAs is limited. In this study, we determined the intracellular and extracellular levels of d-Ser, d-Ala, and d-Asp in cultures of isolated rodent islets exposed to different levels of extracellular glucose. We found that the intracellular levels of the enantiomers demonstrated large variability and, in general, were not affected by extracellular glucose levels. However, significantly lower levels of extracellular d-Ser and d-Ala were observed in the islet media supplemented with 20 mM concentration of glucose compared to the control condition utilizing 3 mM glucose. Glucose-induced oscillations of intracellular free calcium concentration ([Ca2+]i), a proxy for insulin secretion, were modulated by the exogenous application of d-Ser and d-Ala but not by their l-stereoisomers. Our results provide new insights into the roles of d-AAs in the biochemistry and function of pancreatic islets.
Collapse
Affiliation(s)
- Cindy
J. Lee
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Dong-Kyu Lee
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - I-An Wei
- Department
of Chemistry and Biochemistry, Florida State
University, Tallahassee, Florida 32306, United States
| | - Tian A. Qiu
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Stanislav S. Rubakhin
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Michael G. Roper
- Department
of Chemistry and Biochemistry, Florida State
University, Tallahassee, Florida 32306, United States
| | - Jonathan V. Sweedler
- Department
of Chemistry and the Beckman Institute, University of Illinois Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
5
|
Turbitt J, Brennan L, Moffett RC, Flatt PR, Johnson PRV, Tarasov AI, McClenaghan NH. NKCC transport mediates the insulinotropic effects of taurine and other small neutral amino acids. Life Sci 2023; 316:121402. [PMID: 36669678 DOI: 10.1016/j.lfs.2023.121402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
AIMS Despite its high concentration in pancreatic islets of Langerhans and broad range of antihyperglycemic effects, the route facilitating the import of dietary taurine into pancreatic β-cell and mechanisms underlying its insulinotropic activity are unclear. We therefore studied the impact of taurine on beta-cell function, alongside that of other small neutral amino acids, L-alanine and L-proline. MAIN METHODS Pharmacological profiling of insulin secretion was conducted using clonal BRIN BD11 β-cells, the impact of taurine on the metabolic fate of glucose carbons was assessed using NMR and the findings were verified by real-time imaging of Ca2+ dynamics in the cytosol of primary mouse and human islet beta-cells. KEY FINDINGS In our hands, taurine, alanine and proline induced secretory responses that were dependent on the plasma membrane depolarisation, import of Ca2+, homeostasis of K+ and Na+ as well as on cell glycolytic and oxidative metabolism. Taurine shifted the balance between the oxidation and anaplerosis towards the latter, in BRIN BD11 beta-cells. Furthermore, the amino acid signalling was significantly attenuated by inhibition of Na+-K+-Cl- symporter (NKCC). SIGNIFICANCE These data suggest that taurine, like L-alanine and L-proline, acutely induces glucose-dependent insulin-secretory responses by modulating electrogenic Na+ transport, with potential role of intracellular K+ and Cl- in the signal transduction. The acute action delineated would be consistent with antidiabetic potential of dietary taurine supplementation.
Collapse
Affiliation(s)
- Julie Turbitt
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK
| | - Lorraine Brennan
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | - R Charlotte Moffett
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK.
| | - Peter R Flatt
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK.
| | - Paul R V Johnson
- Nuffeld Department of Surgical Sciences, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Headington, OX3 7LE Oxford, UK; Oxford Biomedical Research Centre (OxBRC), UK.
| | - Andrei I Tarasov
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK; Nuffeld Department of Surgical Sciences, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Headington, OX3 7LE Oxford, UK; Oxford Biomedical Research Centre (OxBRC), UK.
| | - Neville H McClenaghan
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK; Department of Life Sciences, Atlantic Technological University, Ash Lane, Sligo, F91 YW50, Ireland.
| |
Collapse
|
6
|
Ezeonwumelu IJ, Mode AM, Magaji UF, Nzoniwu NA, Tangaza MH, Tanimu FI, Dandare SU. Coadministration of L-alanine and L-glutamine ameliorate blood glucose levels, biochemical indices and histological features in alloxan-induced diabetic rats. J Food Biochem 2022; 46:e14420. [PMID: 36125865 DOI: 10.1111/jfbc.14420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 01/13/2023]
Abstract
We evaluated the effects of supplementation of L-alanine and L-glutamine on blood glucose levels and biochemical parameters in alloxan-induced diabetic rat. Forty-nine animals were distributed into seven equal groups. Except for the non-diabetic control, diabetes was induced in all groups by intravenous alloxan injection followed by daily supplementation with amino acids for 14 days. Weight and blood glucose were monitored during supplementation, while biochemical parameters such as liver and renal functions, lipid profile, and antioxidant markers were evaluated post-intervention. A significant increase (p < .05) in weight and decrease in blood glucose were observed in the amino acid(s) treated groups. The supplementation with both amino acids restored important tissue antioxidants, liver and kidney functions and rescued islets cells degeneration. Histopathological examinations of important tissues showed the restoration of alloxan-induced physiopathological changes by the amino acids. Thus, these amino acids might serve as nutraceuticals for the management and treatment of diabetes. PRACTICAL APPLICATIONS: The discovery and production of antidiabetic bioactive compounds are often challenging, and the existing antidiabetic drugs are expensive. Amino acids are key regulators of glucose metabolism, insulin secretion, and insulin sensitivity; thus, they can play a crucial role in alleviating diabetes. Here, we present findings that strongly suggest the potential of pure amino acids (L-alanine and L-glutamine) for the management and treatment of diabetes. We show that these amino acids, when supplemented singly or coadministered can lower blood glucose levels and restore several other biochemical parameters implicated in diabetes. Hence, these cheap amino acids may be consumed as nutraceuticals or food supplements by diabetics for the treatment/management of diabetes. Foods rich in these amino acids may also be consumed as part of the diet of diabetic patients.
Collapse
Affiliation(s)
- Ifeanyi J Ezeonwumelu
- IrsiCaixa - AIDS Research Institute, Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Abduljalil M Mode
- Central Advanced Science Laboratory Complex, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Umar F Magaji
- Department of Chemistry, Biochemistry Unit, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Nnamdi A Nzoniwu
- Abu Dhabi Vocational Education and Training Institute (ADVETI), Mohammed Bin Zayed City, Abu Dhabi, UAE
| | - Muhamad H Tangaza
- Department of Biochemistry, Faculty of Science, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Fatima I Tanimu
- Department of Biochemistry, Faculty of Science, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Shamsudeen U Dandare
- Department of Biochemistry, Faculty of Science, Usmanu Danfodiyo University, Sokoto, Nigeria.,School of Natural and Built Environment, Queen's University Belfast, Belfast, UK
| |
Collapse
|
7
|
Bröer S. Amino acid transporters as modulators of glucose homeostasis. Trends Endocrinol Metab 2022; 33:120-135. [PMID: 34924221 DOI: 10.1016/j.tem.2021.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 11/01/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022]
Abstract
Amino acids modulate glucose homeostasis. Cytosolic levels of amino acids are regulated by amino acid transporters, modulating insulin release, protein synthesis, cell proliferation, cell fate, and metabolism. In β-cells, amino acid transporters modulate incretin-stimulated insulin release. In the liver, amino acid transporters provide glutamine and alanine for gluconeogenesis. Intestinal amino acid transporters facilitate the intake of amino acids causing protein restriction when inactive. Adipocyte development is regulated by amino acid transporters through activation of mechanistic target of rapamycin (mTORC1) and amino acid-related metabolites. The accumulation and metabolism of branched-chain amino acids (BCAAs) in muscle depends on transporters. The integration between amino acid metabolism and transport is critical for the maintenance and function of tissues and cells involved in glucose homeostasis.
Collapse
Affiliation(s)
- Stefan Bröer
- Research School of Biology, Australian National University, Acton 2601, Australia.
| |
Collapse
|
8
|
Arora A, Behl T, Sehgal A, Singh S, Sharma N, Chigurupati S, Kaur R, Bhatia S, Al-Harrasi A, Vargas-De-La-Cruz C, Bungau S. Free fatty acid receptor 1: a ray of hope in the therapy of type 2 diabetes mellitus. Inflammopharmacology 2021; 29:1625-1639. [PMID: 34669065 DOI: 10.1007/s10787-021-00879-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022]
Abstract
Free fatty acid receptor 1 (FFAR1) is a G-protein coupled receptor with prominent expression on pancreatic beta cells, bones, intestinal cells as well as the nerve cells. This receptor mediates a multitude of functions in the body including release of incretins, secretion of insulin as well as sensation of pain. Since FFAR1 causes secretion of insulin and regulates glucose metabolism, efforts were made to unfold its structure followed by discovering agonists for the receptor and the utilization of these agonists in the therapy of type 2 diabetes mellitus. Development of such functional FFAR1 agonists is a necessity because the currently available therapy for type 2 diabetes mellitus has numerous drawbacks, of which, the major one is hypoglycemia. Since the most prominent effect of the FFAR1 agonists is on glucose concentration in the body, so the major research is focused on treating type 2 diabetes mellitus, though the agonists could benefit other metabolic disorders and neurological disorders as well. The agonists developed so far had one major limitation, i.e., hepatotoxicity. Although, the only agonist that could reach phase 3 clinical trials was TAK-875 developed by Takeda Pharmaceuticals but it was also withdrawn due to toxic effects on the liver. Thus, there are numerous agonists for the varied binding sites of the receptor but no drug available yet. There does seem to be a ray of hope in the drugs that target FFAR1 but a lot more efforts towards drug discovery would result in the successful management of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Arpita Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Bhatia
- Natural and Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Celia Vargas-De-La-Cruz
- Faculty of Pharmacy and Biochemistry, Academic Department of Pharmacology, Bromatology and Toxicology, Centro Latinoamericano de Ensenanza e Investigacion en Bacteriologia Alimentaria, Universidad Nacional Mayor de San Marcos, Lima, Peru
- E-Health Research Center, Universidad de Ciencias y Humanidades, Lima, Peru
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
9
|
Physicochemical, Nutritional and In Vitro Antidiabetic Characterisation of Blue Whiting ( Micromesistiuspoutassou) Protein Hydrolysates. Mar Drugs 2021; 19:md19070383. [PMID: 34356808 PMCID: PMC8304566 DOI: 10.3390/md19070383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 01/19/2023] Open
Abstract
Protein hydrolysates from low-value underutilised fish species are potential sources of high-quality dietary protein and health enhancing peptides. Six blue whiting soluble protein hydrolysates (BW-SPH-A_F), generated at industrial scale using different hydrolysis conditions, were assessed in terms of their protein equivalent content, amino acid profile and score and physicochemical properties in addition to their ability to inhibit dipeptidyl peptidase IV (DPP-IV) and stimulate the secretion of insulin from BRIN-BD11 cells. Furthermore, the effect of simulated gastrointestinal digestion (SGID) on the stability of the BW-SPHs and their associated in vitro antidiabetic activity was investigated. The BW-SPHs contained between 70–74% (w/w) protein and all essential and non-essential amino acids. All BW-SPHs mediated DPP-IV inhibitory (IC50: 2.12–2.90 mg protein/mL) and insulin secretory activity (2.5 mg/mL; 4.7 to 6.4-fold increase compared to the basal control (5.6 mM glucose alone)). All BW-SPHs were further hydrolysed during SGID. While the in vitro DPP-IV inhibitory and insulin secretory activity mediated by some BW-SPHs was reduced following SGID, the activity remained high. In general, the insulin secretory activity of the BW-SPHs were 4.5–5.4-fold higher than the basal control following SGID. The BW-SPHs generated herein provide potential for anti-diabetic related functional ingredients, whilst also enhancing environmental and commercial sustainability.
Collapse
|
10
|
Matteo G, Hoyeck MP, Blair HL, Zebarth J, Rick KRC, Williams A, Gagné R, Buick JK, Yauk CL, Bruin JE. Prolonged Low-Dose Dioxin Exposure Impairs Metabolic Adaptability to High-Fat Diet Feeding in Female but Not Male Mice. Endocrinology 2021; 162:bqab050. [PMID: 33693622 PMCID: PMC8101695 DOI: 10.1210/endocr/bqab050] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Human studies consistently show an association between exposure to persistent organic pollutants, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD, aka "dioxin"), and increased diabetes risk. We previously showed that a single high-dose TCDD exposure (20 µg/kg) decreased plasma insulin levels in male and female mice in vivo, but effects on glucose homeostasis were sex-dependent. OBJECTIVE The current study assessed whether prolonged exposure to a physiologically relevant low-dose of TCDD impacts glucose homeostasis and/or the islet phenotype in a sex-dependent manner in chow-fed or high-fat diet (HFD)-fed mice. METHODS Male and female mice were exposed to 20 ng/kg/d TCDD 2×/week for 12 weeks and simultaneously fed standard chow or a 45% HFD. Glucose homeostasis was assessed by glucose and insulin tolerance tests, and glucose-induced plasma insulin levels were measured in vivo. Histological analysis was performed on pancreas from male and female mice, and islets were isolated from females for TempO-Seq transcriptomic analysis. RESULTS Low-dose TCDD exposure did not lead to adverse metabolic consequences in chow-fed male or female mice, or in HFD-fed males. However, TCDD accelerated the onset of HFD-induced hyperglycemia and impaired glucose-induced plasma insulin levels in females. TCDD caused a modest increase in islet area in males but reduced the percent beta cell area within islets in females. TempO-Seq analysis suggested abnormal changes to endocrine and metabolic pathways in female TCDDHFD islets. CONCLUSION Our data suggest that prolonged low-dose TCDD exposure has minimal effects on glucose homeostasis and islet morphology in chow-fed male and female mice but promotes maladaptive metabolic responses in HFD-fed females.
Collapse
Affiliation(s)
- Geronimo Matteo
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Myriam P Hoyeck
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Hannah L Blair
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Julia Zebarth
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Kayleigh R C Rick
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Rémi Gagné
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Julie K Buick
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario K1A 0K9, Canada
- Department of Biology, University of Ottawa, Ontario, KIN 6N5, Canada
| | - Jennifer E Bruin
- Department of Biology & Institute of Biochemistry, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| |
Collapse
|
11
|
Losada-Barragán M. Physiological effects of nutrients on insulin release by pancreatic beta cells. Mol Cell Biochem 2021; 476:3127-3139. [PMID: 33844157 DOI: 10.1007/s11010-021-04146-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Obesity and type 2 diabetes (T2D) are growing health problems associated with a loss of insulin sensitivity. Both conditions arise from a long-term energy imbalance, and frequently, lifestyle measures can be useful in its prevention, including physical activity and a healthy diet. Pancreatic β-cells are determinant nutrient sensors that participate in energetic homeostasis needs. However, when pancreatic β-cells are incapable of secreting enough insulin to counteract the reduced sensitivity, the pathology evolves to an insulin resistance condition. The primary nutrient that stimulates insulin secretion is glucose, but also, there are multiple dietary and hormonal factors influencing that response. Many studies of the physiology of β-cells have highlighted the importance of glucose, fructose, amino acids, and free fatty acids on insulin secretion. The present review summarizes recent research on how β-cells respond to the most abundant nutrients that influence insulin secretion. Taken together, understand the subjacent mechanisms of each nutrient on β-cells can help to unravel the effects of mixed variables and complexity in the context of β-cell pathology.
Collapse
Affiliation(s)
- Monica Losada-Barragán
- Grupo de investigación en Biología celular y funcional e ingeniería de biomoléculas, Universidad Antonio Nariño-Sede Circunvalar. Cra, 3 Este # 47A - 15, Bl 5, Bogotá, Colombia.
| |
Collapse
|
12
|
Zhong C, Zhang F, Yao J, Zhu Y, Zhu N, Zhang Y, Liu H, Gou S, Ni J. Antimicrobial peptides with symmetric structures against multidrug-resistant bacteria while alleviating antimicrobial resistance. Biochem Pharmacol 2021; 186:114470. [PMID: 33610592 DOI: 10.1016/j.bcp.2021.114470] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/26/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022]
Abstract
In response to the dramatically increasing antimicrobial resistance, a series of new symmetric peptides were designed and synthesized in this study by a "WWW" motif as the symmetric center, arginine as the positive charge amino acid and the terminus symmetrically tagged with hydrophobic amino acids. Amongst the new symmetric peptide FRRW (FRRWWWRRF-NH2) presented the highest cell selectivity for bacteria over mammalian cell and exerted excellent antimicrobial potential against a broad of bacteria, especially difficult-to-kill multidrug-resistant strains clinical isolates. FRRW also displayed perfect stability in physiological salt ions and rapid killing speed as well as acted on multiple mechanisms including non-receptor mediated membrane and intra-molecular mechanisms. Importantly, FRRW emerged a low tendency of resistance in contrast to traditional antibiotics ciprofloxacin and gentamicin. What's more, FRRW could resist or alleviate or even reverse the ciprofloxacin- and gentamicin-resistance by changing the permeability of bacterial membrane and inhibiting the efflux pumps of bacteria. Furthermore, FRRW exhibited remarkable effectiveness and higher safety in vivo than polymyxin B. In summary, the new symmetric peptide FRRW was promised to be as a new antimicrobial candidate for overcoming the increasing bacterial resistance.
Collapse
Affiliation(s)
- Chao Zhong
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Fangyan Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jia Yao
- The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yuewen Zhu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Ningyi Zhu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yun Zhang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Hui Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Sanhu Gou
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jingman Ni
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; School of Pharmacy, Lanzhou University, Lanzhou 730000, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao 999078, China.
| |
Collapse
|
13
|
Dandare SU, Ezeonwumelu IJ, Shinkafi TS, Magaji UF, Adio AAI, Ahmad K. L-alanine supplementation improves blood glucose level and biochemical indices in alloxan-induced diabetic rats. J Food Biochem 2020; 45:e13590. [PMID: 33346923 DOI: 10.1111/jfbc.13590] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 11/30/2022]
Abstract
Diabetes is a metabolic disorder whose complications are among the leading cause of death. In this study, the antidiabetic effect of L-alanine was tested in alloxan-induced diabetic rats. Thirty-five male albino Wistar rats were divided into five groups viz; Group I and II: nondiabetic and diabetic controls respectively; Group III and IV: 150 and 300 mg/kg b.w. L-alanine treated, respectively; Group V: glibenclamide (0.5 mg/kg b.w.) treated. Weight and blood glucose were monitored during the study, while liver and kidney functions, lipid profile, and antioxidant markers were examined at the end of the study. The outcomes indicate that 300 mg/kg L-alanine resulted to a significant decrease (p < .05) in weight and blood glucose. L-alanine restored tissue antioxidants, kidney, and liver functions by improving important parameters. Histopathological studies showed the potential of L-alanine in regeneration of the islets of Langerhans. These findings suggest that L-alanine has an alleviating effect on alloxan-induced diabetes. PRACTICAL APPLICATIONS: Several medicinal plants have been tested for their antidiabetic potentials, however, the isolation of the active compounds from these plants for medicinal use is often challenging. Here, we present data that suggests the potential use of a pure and harmless amino acid compound (L-alanine) for the management of diabetes. L-alanine is readily available, cheap and can also be found in many foods we eat. Therefore, L-alanine may be taken by diabetic patients as a food supplement for the treatment/management of diabetes or taken as part of foods rich in the amino acid such as meat, poultry, fish, eggs, and dairy products.
Collapse
Affiliation(s)
- Shamsudeen Umar Dandare
- Department of Biochemistry, Faculty of Science, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Ifeanyi Jude Ezeonwumelu
- IrsiCaixa - AIDS Research Institute, Health Research Institute Germans Trias I Pujol Research Institute, IGTP, Badalona, Spain
| | - Tijjani Salihu Shinkafi
- Department of Biochemistry, Faculty of Science, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Umar Faruk Magaji
- Department of Chemistry, Biochemistry Unit, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | | | - Kasimu Ahmad
- Department of Biochemistry, Faculty of Science, Usmanu Danfodiyo University, Sokoto, Nigeria
| |
Collapse
|
14
|
Razavi M, Primavera R, Kevadiya BD, Wang J, Ullah M, Buchwald P, Thakor AS. Controlled Nutrient Delivery to Pancreatic Islets Using Polydopamine-Coated Mesoporous Silica Nanoparticles. NANO LETTERS 2020; 20:7220-7229. [PMID: 32909757 PMCID: PMC8121116 DOI: 10.1021/acs.nanolett.0c02576] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
In the present study, we created a nanoscale platform that can deliver nutrients to pancreatic islets in a controlled manner. Our platform consists of a mesoporous silica nanoparticle (MSNP), which can be loaded with glutamine (G: an essential amino acid required for islet survival and function). To control the release of G, MSNPs were coated with a polydopamine (PD) layer. With the optimal parameters (0.5 mg/mL and 0.5 h), MSNPs were coated with a layer of PD, which resulted in a delay of G release from MSNPs over 14 d (57.4 ± 4.7% release). Following syngeneic renal subcapsule islet transplantation in diabetic mice, PDG-MSNPs improved the engraftment of islets (i.e., enhanced revascularization and reduced inflammation) as well as their function, resulting in re-establishment of glycemic control. Collectively, our data show that PDG-MSNPs can support transplanted islets by providing them with a controlled and sustained supply of nutrients.
Collapse
Affiliation(s)
- Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States; Biionix (Bionic Materials, Implants & Interfaces) Cluster, Department of Internal Medicine, College of Medicine and Department of Materials Science & Engineering, University of Central Florida, Orlando, Florida 32827, United States
| | - Rosita Primavera
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Bhavesh D Kevadiya
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33136, United States
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Palo Alto, California 94304, United States
| |
Collapse
|
15
|
Chou S, Li Q, Nina Z, Shang L, Li J, Li J, Wang Z, Shan A. Peptides With Triplet-Tryptophan-Pivot Promoted Pathogenic Bacteria Membrane Defects. Front Microbiol 2020; 11:537. [PMID: 32328042 PMCID: PMC7160233 DOI: 10.3389/fmicb.2020.00537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/12/2020] [Indexed: 02/05/2023] Open
Abstract
Development of probiotic-ineffective antimicrobial peptides (AMPs)-based coatings that can kill pathogenic bacteria at low concentrations but are essentially harmless (even high concentrations) to probiotic organisms is a relatively new trend for therapy against GI tract infections. In this study, a series of triplet-tryptophan-pivot peptides with various hydrophilic amino acids was constructed. One AMP in particular, S7, showed bactericidal activity against Staphylococcus epidermidis, Pseudomonas aeruginosa, Escherichia coli and antibiotic-resistant Staphylococcus aureus, yet was shown to be harmless to Lactobacillus rhamnosus, a key GI tract commensal. Furthermore, antibacterial mechanism assays, drug resistance assays, and mouse model tests suggested that S7 was useful in a clinical setting as it proved to significantly reduce bacterial load and cytokines (TNF-α, IL-6; P < 0.05) with a low probability of resistance via bacterial membrane physical destruction and formation of intracellular ROS. Combined, the results show that a triplet-tryptophan-pivot peptide containing a pair of serine residues was an excellent pathogen-selective candidate for medical devices and was potentially useful in food preservation, crop protection, and human health.
Collapse
Affiliation(s)
- Shuli Chou
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Qiuke Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zaitseva Nina
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Lu Shang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Jiawei Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Jinze Li
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Zhihua Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Anshan Shan
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
16
|
Synthesis and evaluation of novel peptidomimetics bearing p-aminobenzoic acid moiety as potential antidiabetic agents. Future Med Chem 2020; 12:991-1013. [PMID: 32208864 DOI: 10.4155/fmc-2018-0372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Search for a new class of potential antidiabetic agents. Methodology: A series of novel peptidomimetics bearing the p-aminobenzoic acid moiety (TM3-TM6) were designed and synthesized. For all synthetic target molecules, the peroxisome proliferator response element (PPRE) activated activities have been evaluated and the toxicity were computed. Results & discussion: 46 new p-aminobenzoic acid derivatives have been characterized by 1H NMR, 13C NMR and high-resolution mass spectrometry (HRMS). The results of in vitro PPRE-activated activity, molecular docking study and toxicity prediction revealed that these compounds had potential antidiabetic activities and low toxicity. In particular compound 3b had up to 87% PPRE-activated activity compared with pioglitazone. This discovery may provide new insights for finding novel PPRE lead compound.
Collapse
|
17
|
Harnedy-Rothwell PA, McLaughlin CM, O'Keeffe MB, Le Gouic AV, Allsopp PJ, McSorley EM, Sharkey S, Whooley J, McGovern B, O'Harte FPM, FitzGerald RJ. Identification and characterisation of peptides from a boarfish (Capros aper) protein hydrolysate displaying in vitro dipeptidyl peptidase-IV (DPP-IV) inhibitory and insulinotropic activity. Food Res Int 2020; 131:108989. [PMID: 32247474 DOI: 10.1016/j.foodres.2020.108989] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 10/30/2019] [Accepted: 01/06/2020] [Indexed: 02/08/2023]
Abstract
Twenty-two novel dipeptidyl peptidase-IV (DPP-IV) inhibitory peptides (with IC50 values <200 µM) and fifteen novel insulinotropic peptides were identified in a boarfish protein hydrolysate generated at semi-pilot scale using Alcalase 2.4L and Flavourzyme 500L. This was achieved by bioassay-driven semi-preparative reverse phase-high performance liquid chromatography fractionation, liquid chromatography-mass spectrometry and confirmatory studies with synthetic peptides. The most potent DPP-IV inhibitory peptide (IPVDM) had a DPP-IV half maximal inhibitory concentration (IC50) value of 21.72 ± 1.08 µM in a conventional in vitro and 44.26 ± 0.65 µM in an in situ cell-based (Caco-2) DPP-IV inhibition assay. Furthermore, this peptide stimulated potent insulin secretory activity (1.6-fold increase compared to control) from pancreatic BRIN-BD11 cells grown in culture. The tripeptide IPV exhibited potent DPP-IV inhibitory activity (IC50: 5.61 ± 0.20 µM) comparable to that reported for the known DPP-IV inhibitor IPI (IC50: 3.20 µM). Boarfish proteins contain peptide sequences with potential to play a role in glycaemic management in vivo.
Collapse
Affiliation(s)
| | - Chris M McLaughlin
- School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry, Northern Ireland, United Kingdom
| | - Martina B O'Keeffe
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Aurélien V Le Gouic
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Philip J Allsopp
- School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry, Northern Ireland, United Kingdom
| | - Emeir M McSorley
- School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry, Northern Ireland, United Kingdom
| | - Shaun Sharkey
- School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry, Northern Ireland, United Kingdom
| | - Jason Whooley
- Bio-Marine Ingredients Ireland Ltd., Lough Egish Food Park, Castleblaney, Co. Monaghan, Ireland
| | - Brian McGovern
- Bio-Marine Ingredients Ireland Ltd., Lough Egish Food Park, Castleblaney, Co. Monaghan, Ireland
| | - Finbarr P M O'Harte
- School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry, Northern Ireland, United Kingdom
| | | |
Collapse
|
18
|
Carlessi R, Rowlands J, Ellison G, Helena de Oliveira Alves H, Newsholme P, Mamotte C. Glutamine deprivation induces metabolic adaptations associated with beta cell dysfunction and exacerbate lipotoxicity. Mol Cell Endocrinol 2019; 491:110433. [PMID: 31018148 DOI: 10.1016/j.mce.2019.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 11/17/2022]
Abstract
Studies have reported that plasma glutamine is reduced in type 2 diabetes (T2D) patients. Glutamine supplementation improves glycaemic control, however the mechanisms are unclear. Here, we evaluated in vitro the pancreatic beta cell bioenergetic and insulin secretory responses to various levels of glutamine availability, or treatment in the presence of an inhibitor of intracellular glutamine metabolism. The impact of glutamine deprivation to the pathological events induced by the saturated fatty acid palmitate was also investigated. Glutamine deprivation induced a reduction in mitochondrial respiration and increase in glucose uptake and utilization. This phenotype was accompanied by impairment in beta cell function, as demonstrated by diminished insulin production and secretion, and activation of the unfolded protein response pathway. Palmitate led to insulin secretory dysfunction, loss of viability and apoptosis. Importantly, glutamine deprivation significantly exacerbated these phenotypes, suggesting that low glutamine levels could participate in the process of beta cell dysfunction in T2D.
Collapse
Affiliation(s)
- Rodrigo Carlessi
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, Western Australia, 6845, Australia.
| | - Jordan Rowlands
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, Western Australia, 6845, Australia
| | - Gaewyn Ellison
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, Western Australia, 6845, Australia
| | | | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, Western Australia, 6845, Australia.
| | - Cyril Mamotte
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, Western Australia, 6845, Australia.
| |
Collapse
|
19
|
Yang C, Hao R, Du X, Wang Q, Deng Y, Sun R. Response to different dietary carbohydrate and protein levels of pearl oysters (Pinctada fucata martensii) as revealed by GC-TOF/MS-based metabolomics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 650:2614-2623. [PMID: 30373048 DOI: 10.1016/j.scitotenv.2018.10.023] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 06/08/2023]
Abstract
Land-based culturing can avoid the effects of environmental pollution and natural disasters, thus ensuring food safety for shellfish. However, food availability, in this case, is limited. To achieve the optimum balance of dietary carbohydrates and proteins and explore the mechanisms behind the phenomenon, we formulated five isoenergetic and isolipidic diets (C30P40, C35P35, C40P30, C45P25, and C50P20) with different levels of carbohydrates (C) and proteins (P). There were five experimental groups (C30P40, C35P35, C40P30, C45P25, and C50P20) and two control groups (CG1 and CG2). CG1 was fed with mixed powders of yeast and Chlorella sp., and CG2 was cultured in natural sea. After 60-day feeding, the highest rates of survival and absolute growth appeared in C45P25. C45P25 exhibited significantly higher activities of amylase, protease, alkaline phosphatase, acid phosphatase, superoxide dismutase, catalase, glutathione peroxidase, and phenoloxidase and significantly lower malondialdehyde content than C30P40, C35P35, C40P30, C50P20, and CG1. No significant differences were observed between C45P25 and CG2. Furthermore, the total antioxidant capacity of the pearl oysters in C45P25 was significantly higher than that in C30P40, C35P35, C40P30, and C50P20. On the basis of these results, the optimal balance of proteins and carbohydrates for pearl oysters was the C45P25 diet. Metabolomics-based profiling of the pearl oysters fed with high-carbohydrate/low-protein diet (C45P25) and low-carbohydrate/high-protein diet (C30P40) revealed 80 significantly different metabolites (VIP > 1 and P < 0.05). Furthermore, integrated key metabolic pathway analysis showed that C45P25 regulated starch and sucrose metabolism, alanine, aspartate and glutamate metabolism and glycine, serine and threonine metabolism to meet the energy demand and increase the glucogenic amino acid, thereby promoting protein synthesis and reducing fatty acid β-oxidation in comparison with C30P40. This finding helps elucidate the underlying mechanisms leading to the high-carbohydrate/low-protein diet characteristic of the optimal dietary carbohydrate and protein levels of P. f. martensii.
Collapse
Affiliation(s)
- Chuangye Yang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Ruijuan Hao
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaodong Du
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang 524088, China.
| | - Qingheng Wang
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang 524088, China
| | - Yuewen Deng
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; Pearl Breeding and Processing Engineering Technology Research Centre of Guangdong Province, Zhanjiang 524088, China
| | - Ruijiao Sun
- Zhejiang Hengxing Food Co., Ltd., Jiaxing 314100, China
| |
Collapse
|
20
|
Tapadia M, Carlessi R, Johnson S, Utikar R, Newsholme P. Lupin seed hydrolysate promotes G-protein-coupled receptor, intracellular Ca 2+ and enhanced glycolytic metabolism-mediated insulin secretion from BRIN-BD11 pancreatic beta cells. Mol Cell Endocrinol 2019; 480:83-96. [PMID: 30347229 DOI: 10.1016/j.mce.2018.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/14/2022]
Abstract
Lupin seed proteins have been reported to exhibit hypoglycaemic effects in animals and humans following oral administration, however little is known about its mechanism of action. This study investigated the signalling pathway(s) responsible for the insulinotropic effect of the hydrolysate obtained from lupin (Lupinus angustifolius L.) seed extracts utilizing BRIN-BD11 β-cells. The extract was treated with digestive enzymes to give a hydrolysate rich in biomolecules ≤7 kDa. Cells exhibited hydrolysate induced dose-dependent stimulation of insulin secretion and enhanced intracellular Ca2+ and glucose metabolism. The stimulatory effect of the hydrolysate was potentiated by depolarizing concentrations of KCl and was blocked by inhibitors of the ATP sensitive K+ channel, Gαq protein, phospholipase C (PLC) and protein kinase C (PKC). These findings reveal a novel mechanism for lupin hydrolysate stimulated insulin secretion via Gαq mediated signal transduction (Gαq/PLC/PKC) in the β-cells. Thus, lupin hydrolysates may have potential for nutraceutical treatment in type 2 diabetes.
Collapse
Affiliation(s)
- Mrunmai Tapadia
- Western Australia School of Mines (WASM): Minerals, Energy and Chemical Engineering, Curtin University, Perth, WA, 6102, Australia
| | - Rodrigo Carlessi
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute Biosciences, Curtin University, Perth, WA, 6102, Australia.
| | - Stuart Johnson
- School of Molecular and Life Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6845, Australia
| | - Ranjeet Utikar
- Western Australia School of Mines (WASM): Minerals, Energy and Chemical Engineering, Curtin University, Perth, WA, 6102, Australia
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute Biosciences, Curtin University, Perth, WA, 6102, Australia.
| |
Collapse
|
21
|
Lin W, Liu Z, Zheng X, Chen M, Gao D, Tian Z. High-salt diet affects amino acid metabolism in plasma and muscle of Dahl salt-sensitive rats. Amino Acids 2018; 50:1407-1414. [DOI: 10.1007/s00726-018-2615-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/03/2018] [Indexed: 10/28/2022]
|
22
|
Ramadhan AH, Nawas T, Zhang X, Pembe WM, Xia W, Xu Y. Purification and identification of a novel antidiabetic peptide from Chinese giant salamander (Andrias davidianus) protein hydrolysate against α-amylase and α-glucosidase. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2018. [DOI: 10.1080/10942912.2017.1354885] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Abuubakar Hassan Ramadhan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Tazbidul Nawas
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Xiaowei Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Warda Mwinyi Pembe
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Wenshui Xia
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| | - Yanshun Xu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, P. R. China
| |
Collapse
|
23
|
Harnedy PA, Parthsarathy V, McLaughlin CM, O'Keeffe MB, Allsopp PJ, McSorley EM, O'Harte FPM, FitzGerald RJ. Atlantic salmon (Salmo salar) co-product-derived protein hydrolysates: A source of antidiabetic peptides. Food Res Int 2018; 106:598-606. [PMID: 29579965 DOI: 10.1016/j.foodres.2018.01.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/26/2022]
Abstract
Large quantities of low-value protein rich co-products, such as salmon skin and trimmings, are generated annually. These co-products can be upgraded to high-value functional ingredients. The aim of this study was to assess the antidiabetic potential of salmon skin gelatin and trimmings-derived protein hydrolysates in vitro. The gelatin hydrolysate generated with Alcalase 2.4L and Flavourzyme 500L exhibited significantly higher (p < 0.001) insulin and GLP-1 secretory activity from pancreatic BRIN-BD11 and enteroendocrine GLUTag cells, respectively, when tested at 2.5 mg/mL compared to hydrolysates generated with Alcalase 2.4L or Promod 144MG. The gelatin hydrolysate generated with Alcalase 2.4L and Flavourzyme 500L showed significantly more potent (p < 0.01) DPP-IV inhibitory activity than those generated with Alcalase 2.4L or Promod 144MG. No significant difference was observed in the insulinotropic activity mediated by any of the trimmings-derived hydrolysates when tested at 2.5 mg/mL. However, the trimmings hydrolysate generated with Alcalase 2.4L and Flavourzyme 500L exhibited significantly higher DPP-IV inhibitory (p < 0.05:Alcalase 2.4L and p < 0.01:Promod 144MG) and GLP-1 (p < 0.001, 2.5 mg/mL) secretory activity than those generated with Alcalase 2.4L or Promod 144MG. The salmon trimmings hydrolysate generated with Alcalase 2.4L and Flavourzyme 500L when subjected to simulated gastrointestinal digestion (SGID) was shown to retain its GLP-1 secretory and DPP-IV inhibitory activities, in addition to improving its insulin secretory activity. However, the gelatin hydrolysate generated with Alcalase 2.4L and Flavourzyme 500L was shown to lose GLP-1 secretory activity following SGID. A significant increase in membrane potential (p < 0.001) and intracellular calcium (p < 0.001) by both co-product hydrolysates generated with Alcalase 2.4L and Flavourzyme 500L suggest that both hydrolysates mediate their insulinotropic activity through the KATP channel-dependent pathway. Additionally, by stimulating a significant increase in intracellular cAMP release (p < 0.05) it is likely that the trimmings-derived hydrolysate may also mediate insulin secretion through the protein kinase A pathway. The results presented herein demonstrate that salmon co-product hydrolysates exhibit promising in vitro antidiabetic activity.
Collapse
Affiliation(s)
- Pádraigín A Harnedy
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Vadivel Parthsarathy
- The SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry BT52 1SA, Northern Ireland.
| | - Chris M McLaughlin
- The SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry BT52 1SA, Northern Ireland.
| | - Martina B O'Keeffe
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Philip J Allsopp
- Northern Ireland Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry BT52 1SA, Northern Ireland.
| | - Emeir M McSorley
- Northern Ireland Centre for Food and Health, School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry BT52 1SA, Northern Ireland.
| | - Finbarr P M O'Harte
- The SAAD Centre for Pharmacy & Diabetes, School of Biomedical Sciences, Ulster University, Coleraine, Co. Derry BT52 1SA, Northern Ireland.
| | | |
Collapse
|
24
|
Harnedy PA, Parthsarathy V, McLaughlin CM, O'Keeffe MB, Allsopp PJ, McSorley EM, O'Harte FP, FitzGerald RJ. Blue whiting (Micromesistius poutassou) muscle protein hydrolysate with in vitro and in vivo antidiabetic properties. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.10.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
25
|
Faleo G, Russ HA, Wisel S, Parent AV, Nguyen V, Nair GG, Freise JE, Villanueva KE, Szot GL, Hebrok M, Tang Q. Mitigating Ischemic Injury of Stem Cell-Derived Insulin-Producing Cells after Transplant. Stem Cell Reports 2017; 9:807-819. [PMID: 28803916 PMCID: PMC5599226 DOI: 10.1016/j.stemcr.2017.07.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/21/2022] Open
Abstract
The advent of large-scale in vitro differentiation of human stem cell-derived insulin-producing cells (SCIPC) has brought us closer to treating diabetes using stem cell technology. However, decades of experiences from islet transplantation show that ischemia-induced islet cell death after transplant severely limits the efficacy of the therapy. It is unclear to what extent human SCIPC are susceptible to ischemia. In this study, we show that more than half of SCIPC die shortly after transplantation. Nutrient deprivation and hypoxia acted synergistically to kill SCIPC in vitro. Amino acid supplementation rescued SCIPC from nutrient deprivation, likely by providing cellular energy. Generating SCIPC under physiological oxygen tension of 5% conferred hypoxia resistance without affecting their differentiation or function. A two-pronged strategy of physiological oxygen acclimatization during differentiation and amino acid supplementation during transplantation significantly improved SCIPC survival after transplant. Stem cell-derived insulin-producing cells (SCIPC) are susceptible to ischemic injury Amino acid supplementation prevents nutrient-deprivation-induced SCIPC death Generation of SCIPC at physiological oxygen levels protects them against hypoxia Both strategies combined preserve SCIPC graft viability in vivo upon transplant
Collapse
Affiliation(s)
- Gaetano Faleo
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Holger A Russ
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA; Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Steven Wisel
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Audrey V Parent
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vinh Nguyen
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Gopika G Nair
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jonathan E Freise
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Karina E Villanueva
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Gregory L Szot
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Matthias Hebrok
- UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA; UCSF Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
26
|
GLP-1 receptor signalling promotes β-cell glucose metabolism via mTOR-dependent HIF-1α activation. Sci Rep 2017; 7:2661. [PMID: 28572610 PMCID: PMC5454020 DOI: 10.1038/s41598-017-02838-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/19/2017] [Indexed: 02/05/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) promotes insulin secretion from pancreatic β-cells in a glucose dependent manner. Several pathways mediate this action by rapid, kinase phosphorylation-dependent, but gene expression-independent mechanisms. Since GLP-1-induced insulin secretion requires glucose metabolism, we aimed to address the hypothesis that GLP-1 receptor (GLP-1R) signalling can modulate glucose uptake and utilization in β-cells. We have assessed various metabolic parameters after short and long exposure of clonal BRIN-BD11 β-cells and rodent islets to the GLP-1R agonist Exendin-4 (50 nM). Here we report for the first time that prolonged stimulation of the GLP-1R for 18 hours promotes metabolic reprogramming of β-cells. This is evidenced by up-regulation of glycolytic enzyme expression, increased rates of glucose uptake and consumption, as well as augmented ATP content, insulin secretion and glycolytic flux after removal of Exendin-4. In our model, depletion of Hypoxia-Inducible Factor 1 alpha (HIF-1α) impaired the effects of Exendin-4 on glucose metabolism, while pharmacological inhibition of Phosphoinositide 3-kinase (PI3K) or mTOR completely abolished such effects. Considering the central role of glucose catabolism for stimulus-secretion coupling in β-cells, our findings suggest that chronic GLP-1 actions on insulin secretion include elevated β-cell glucose metabolism. Moreover, our data reveal novel aspects of GLP-1 stimulated insulin secretion involving de novo gene expression.
Collapse
|
27
|
Xia EQ, Zhu SS, He MJ, Luo F, Fu CZ, Zou TB. Marine Peptides as Potential Agents for the Management of Type 2 Diabetes Mellitus-A Prospect. Mar Drugs 2017; 15:md15040088. [PMID: 28333091 PMCID: PMC5408234 DOI: 10.3390/md15040088] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 01/09/2023] Open
Abstract
An increasing prevalence of diabetes is known as a main risk for human health in the last future worldwide. There is limited evidence on the potential management of type 2 diabetes mellitus using bioactive peptides from marine organisms, besides from milk and beans. We summarized here recent advances in our understanding of the regulation of glucose metabolism using bioactive peptides from natural proteins, including regulation of insulin-regulated glucose metabolism, such as protection and reparation of pancreatic β-cells, enhancing glucose-stimulated insulin secretion and influencing the sensitivity of insulin and the signaling pathways, and inhibition of bioactive peptides to dipeptidyl peptidase IV, α-amylase and α-glucosidase activities. The present paper tried to understand the underlying mechanism involved and the structure characteristics of bioactive peptides responsible for its antidiabetic activities to prospect the utilization of rich marine organism proteins.
Collapse
Affiliation(s)
- En-Qin Xia
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Shan-Shan Zhu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Min-Jing He
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Fei Luo
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Cheng-Zhan Fu
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Tang-Bin Zou
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
28
|
Comar JF, de Oliveira DS, Bracht L, Kemmelmeier FS, Peralta RM, Bracht A. The Metabolic Responses to L-Glutamine of Livers from Rats with Diabetes Types 1 and 2. PLoS One 2016; 11:e0160067. [PMID: 27490892 PMCID: PMC4973899 DOI: 10.1371/journal.pone.0160067] [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] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/12/2016] [Indexed: 11/18/2022] Open
Abstract
There are several claims about the beneficial effects of supplementing L-glutamine to both type 1 and type 2 diabetes. The purpose of the present study was to provide detailed knowledge about the fate of this amino acid in the liver, the first organ that receives the compound when ingested orally. The study was done using the isolated perfused rat liver, an experimental system that preserves the microcirculation of the organ and that allows to measured several parameters during steady-state and pre steady-state conditions. L-Glutamine was infused in the portal vein (5 mM) and several parameters were monitored. Livers from type 1 diabetic rats showed an accelerated response to L-glutamine infusion. In consequence of this accelerated response livers from type 1 diabetic rats presented higher rates of ammonia, urea, glucose and lactate output during the first 25–30 minutes following L-glutamine infusion. As steady-state conditions approached, however, the difference between type 1 diabetes and control livers tended to disappear. Measurement of the glycogen content over a period of 100 minutes revealed that, excepting the initial phase of the L-glutamine infusion, the increased glucose output in livers from type 1 diabetic rats was mainly due to accelerated glycogenolysis. Livers from type 2 diabetic rats behaved similarly to control livers with no accelerated glucose output but with increased L-alanine production. L-Alanine is important for the pancreatic β-cells and from this point of view the oral intake of L-glutamine can be regarded as beneficial. Furthermore, the lack of increased glucose output in livers from type 2 diabetic rats is consistent with observations that even daily L-glutamine doses of 30 g do not increase the glycemic levels in well controlled type 2 diabetes patients.
Collapse
Affiliation(s)
| | | | - Livia Bracht
- Laboratory of Liver Metabolism, University of Maringá, 87020900 Maringá, Brazil
| | | | | | - Adelar Bracht
- Laboratory of Liver Metabolism, University of Maringá, 87020900 Maringá, Brazil
- * E-mail:
| |
Collapse
|
29
|
Nongonierma AB, FitzGerald RJ. Milk proteins as a source of tryptophan-containing bioactive peptides. Food Funct 2016; 6:2115-27. [PMID: 26027501 DOI: 10.1039/c5fo00407a] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Tryptophan (W) is an essential amino acid which is primarily required for protein synthesis. It also acts as a precursor of key biomolecules for human health (serotonin, melatonin, tryptamine, niacin, nicotinamide adenine dinucleotide (NAD), phosphorylated NAD (NADP), quinolinic acid, kynureric acid, etc.). Among dietary proteins, milk proteins are particularly rich in W. W residues within milk proteins may be released by proteolytic/peptidolytic enzymes either as a free amino acid or as part of peptide sequences. Different W-containing peptides originating from milk proteins have been shown in vitro to display a wide range of bioactivities such as angiotensin converting enzyme (ACE) inhibition along with antioxidant, antidiabetic and satiating related properties. Free W has been shown in certain instances to have an effect on cognition and the aforementioned bioactive properties. However, a higher bioactive potency has generally been observed with specific W-containing peptides compared to free W. Since W is thermolabile, the impact of processing on the stability of W-containing peptides needs to be considered. Milk protein-derived W-containing peptides may have significant potential as natural health promoting agents in humans.
Collapse
Affiliation(s)
- Alice B Nongonierma
- Department of Life Sciences and Food for Health Ireland (FHI), University of Limerick, Castletroy, Limerick, Ireland.
| | | |
Collapse
|
30
|
Power-Grant O, Bruen C, Brennan L, Giblin L, Jakeman P, FitzGerald RJ. In vitro bioactive properties of intact and enzymatically hydrolysed whey protein: targeting the enteroinsular axis. Food Funct 2016; 6:972-80. [PMID: 25666373 DOI: 10.1039/c4fo00983e] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Enzymatically hydrolysed milk proteins have a variety of biofunctional effects some of which may be beneficial in the management of type 2 diabetes mellitus. The purpose of this study was to evaluate the effect of commercially available intact and hydrolysed whey protein ingredients (DH 32, DH 45) on markers of the enteroinsular axis (glucagon like peptide-1 secretion, dipeptidyl peptidase IV inhibition, insulin secretion and antioxidant activity) before and after simulated gastrointestinal digestion (SGID). A whey protein hydrolysate, DH32, significantly enhanced (P < 0.05) insulin secretion from BRIN BD11 β-cells compared to the positive control (16.7 mM glucose and 10 mM Ala). The whey protein hydrolysates inhibited dipeptidyl peptidase IV activity, yielding half maximal inhibitory concentration values (IC50) of 1.5 ± 0.1 and 1.1 ± 0.1 mg mL(-1) for the DH 32 and DH 45, samples respectively, and were significantly more potent than the intact whey (P < 0.05). Enzymatic hydrolysis of whey protein significantly enhanced (P < 0.05) its antioxidant activity compared to intact whey, as measured by the oxygen radical absorbance capacity assay (ORAC). This antioxidant activity was maintained (DH 32, P > 0.05) or enhanced (DH 45, P < 0.05) following SGID. Intact whey stimulated GLP-1 secretion from enteroendocrine cells compared to vehicle control (P < 0.05). This data confirm that whey proteins and peptides can act through multiple targets within the enteroinsular axis and as such may have glucoregulatory potential.
Collapse
Affiliation(s)
- O Power-Grant
- Department of Life Sciences, University of Limerick, Limerick, Ireland.
| | | | | | | | | | | |
Collapse
|
31
|
Park S, Sadanala KC, Kim EK. A Metabolomic Approach to Understanding the Metabolic Link between Obesity and Diabetes. Mol Cells 2015; 38:587-96. [PMID: 26072981 PMCID: PMC4507023 DOI: 10.14348/molcells.2015.0126] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 12/19/2022] Open
Abstract
Obesity and diabetes arise from an intricate interplay between both genetic and environmental factors. It is well recognized that obesity plays an important role in the development of insulin resistance and diabetes. Yet, the exact mechanism of the connection between obesity and diabetes is still not completely understood. Metabolomics is an analytical approach that aims to detect and quantify small metabolites. Recently, there has been an increased interest in the application of metabolomics to the identification of disease biomarkers, with a number of well-known biomarkers identified. Metabolomics is a potent approach to unravel the intricate relationships between metabolism, obesity and progression to diabetes and, at the same time, has potential as a clinical tool for risk evaluation and monitoring of disease. Moreover, metabolomics applications have revealed alterations in the levels of metabolites related to obesity-associated diabetes. This review focuses on the part that metabolomics has played in elucidating the roles of metabolites in the regulation of systemic metabolism relevant to obesity and diabetes. It also explains the possible metabolic relation and association between the two diseases. The metabolites with altered profiles in individual disorders and those that are specifically and similarly altered in both disorders are classified, categorized and summarized.
Collapse
Affiliation(s)
- Seokjae Park
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 711-873,
Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu 711-873,
Korea
| | - Krishna Chaitanya Sadanala
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu 711-873,
Korea
| | - Eun-Kyoung Kim
- Department of Brain & Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 711-873,
Korea
- Neurometabolomics Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu 711-873,
Korea
| |
Collapse
|
32
|
Kaufman BA, Li C, Soleimanpour SA. Mitochondrial regulation of β-cell function: maintaining the momentum for insulin release. Mol Aspects Med 2015; 42:91-104. [PMID: 25659350 PMCID: PMC4404204 DOI: 10.1016/j.mam.2015.01.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 01/29/2015] [Accepted: 01/29/2015] [Indexed: 01/15/2023]
Abstract
All forms of diabetes share the common etiology of insufficient pancreatic β-cell function to meet peripheral insulin demand. In pancreatic β-cells, mitochondria serve to integrate the metabolism of exogenous nutrients into energy output, which ultimately leads to insulin release. As such, mitochondrial dysfunction underlies β-cell failure and the development of diabetes. Mitochondrial regulation of β-cell function occurs through many diverse pathways, including metabolic coupling, generation of reactive oxygen species, maintenance of mitochondrial mass, and through interaction with other cellular organelles. In this chapter, we will focus on the importance of enzymatic regulators of mitochondrial fuel metabolism and control of mitochondrial mass to pancreatic β-cell function, describing how defects in these pathways ultimately lead to diabetes. Furthermore, we will examine the factors responsible for mitochondrial biogenesis and degradation and their roles in the balance of mitochondrial mass in β-cells. Clarifying the causes of β-cell mitochondrial dysfunction may inform new approaches to treat the underlying etiologies of diabetes.
Collapse
Affiliation(s)
- Brett A Kaufman
- Division of Cardiology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Changhong Li
- Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott A Soleimanpour
- Division of Metabolism, Endocrinology & Diabetes and Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Fang J, Wang W, Sun S, Wang Y, Li Q, Lu X, Hao Z, Zhang Y. A urine metabonomics study of chronic renal failure and intervention effects of total aglycone extracts of Scutellaria baicalensis in 5/6 nephrectomy rats. RSC Adv 2015. [DOI: 10.1039/c5ra12710f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This study was to clarify the pathogenesis of CRF and action mechanism of TAES.
Collapse
Affiliation(s)
- Junwei Fang
- Center for Traditional Chinese Medicine and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Wenyu Wang
- Center for Traditional Chinese Medicine and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
- MacroStat (China) Clinical Research Co., Ltd
| | - Shujun Sun
- Center for Traditional Chinese Medicine and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Yang Wang
- Center for Traditional Chinese Medicine and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Qianhua Li
- Center for Traditional Chinese Medicine and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| | - Xiong Lu
- Experiment Center for Science and Technology
- Shanghai University of Traditional
- Chinese Medicine
- Shanghai 201203
- China
| | - Zhihui Hao
- Laboratories of Biological Pharmaceutical
- College of Chemical and Pharmaceutical Sciences
- Qingdao Agricultural University
- Qingdao
- China
| | - Yongyu Zhang
- Center for Traditional Chinese Medicine and Systems Biology
- Shanghai University of Traditional Chinese Medicine
- Shanghai 201203
- China
| |
Collapse
|
34
|
Cetik S, Rzajeva A, Malaisse WJ, Sener A. Effect of cytochalasin B on 3-O-[ 14C]-methyl-D-glucose or D-[U- 14C]glucose handling by BRIN-BD11 cells. Biomed Rep 2014; 2:513-516. [PMID: 24944798 DOI: 10.3892/br.2014.269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 02/26/2014] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate the effects of cytochalasin B (20 μM) on the uptake of 3-O-[14C]-methyl-D-glucose or D-[U-14C]glucose (8.3 mM each) by BRIN-BD11 cells. Taking into account the distribution space of tritiated water (3HOH), which was unexpectedly increased shortly after exposure of the cells to cytochalasin B and then progressively returned to its control values, and that of L-[1-14C]glucose, used as an extracellular marker, it was demonstrated that cytochalasin B caused a modest, but significant inhibition of the uptake of D-glucose and its non-metabolized analog by the BRIN-BD11 cells. These findings resemble those observed in acinar or ductal cells of the rat submaxillary gland and displayed a relative magnitude comparable to that found for the inhibition of D-glucose metabolism by cytochalasin B in purified pancreatic islet B cells. These findings reinforce the view that the primary site of action of cytochalasin B is located at the level of the plasma membrane.
Collapse
Affiliation(s)
- Sibel Cetik
- Laboratory of Experimental Hormonology, Brussels Free University, Brussels B-1070, Belgium
| | - Aigun Rzajeva
- Laboratory of Experimental Hormonology, Brussels Free University, Brussels B-1070, Belgium
| | - Willy J Malaisse
- Laboratory of Experimental Hormonology, Brussels Free University, Brussels B-1070, Belgium
| | - Abdullah Sener
- Laboratory of Experimental Hormonology, Brussels Free University, Brussels B-1070, Belgium
| |
Collapse
|
35
|
Abstract
Pancreatic β-cell function is of critical importance in the regulation of fuel homoeostasis, and metabolic dysregulation is a hallmark of diabetes mellitus (DM). The β-cell is an intricately designed cell type that couples metabolism of dietary sources of carbohydrates, amino acids and lipids to insulin secretory mechanisms, such that insulin release occurs at appropriate times to ensure efficient nutrient uptake and storage by target tissues. However, chronic exposure to high nutrient concentrations results in altered metabolism that impacts negatively on insulin exocytosis, insulin action and may ultimately lead to development of DM. Reduced action of insulin in target tissues is associated with impairment of insulin signalling and contributes to insulin resistance (IR), a condition often associated with obesity and a major risk factor for DM. The altered metabolism of nutrients by insulin-sensitive target tissues (muscle, adipose tissue and liver) can result in high circulating levels of glucose and various lipids, which further impact on pancreatic β-cell function, IR and progression of the metabolic syndrome. Here, we have considered the role played by the major nutrient groups, carbohydrates, amino acids and lipids, in mediating β-cell insulin secretion, while also exploring the interplay between amino acids and insulin action in muscle. We also focus on the effects of altered lipid metabolism in adipose tissue and liver resulting from activation of inflammatory processes commonly observed in DM pathophysiology. The aim of this review is to describe commonalities and differences in metabolism related to insulin secretion and action, pertinent to the development of DM.
Collapse
Affiliation(s)
- Philip Newsholme
- School of Biomedical SciencesCHIRI Biosciences Research Precinct, Curtin University, GPO Box U1987, Perth, Western Australia, Australia
| | - Vinicius Cruzat
- School of Biomedical SciencesCHIRI Biosciences Research Precinct, Curtin University, GPO Box U1987, Perth, Western Australia, Australia
| | - Frank Arfuso
- School of Biomedical SciencesCHIRI Biosciences Research Precinct, Curtin University, GPO Box U1987, Perth, Western Australia, Australia
| | - Kevin Keane
- School of Biomedical SciencesCHIRI Biosciences Research Precinct, Curtin University, GPO Box U1987, Perth, Western Australia, Australia
| |
Collapse
|
36
|
Abstract
Regulation of metabolic fuel homeostasis is a critical function of β-cells, which are located in the islets of Langerhans of the animal pancreas. Impairment of this β-cell function is a hallmark of pancreatic β-cell failure and may lead to development of type 2 diabetes mellitus. β-Cells are essentially "fuel sensors" that monitor and react to elevated nutrient load by releasing insulin. This response involves metabolic activation and generation of metabolic coupling factors (MCFs) that relay the nutrient signal throughout the cell and induce insulin biosynthesis and secretion. Glucose is the most important insulin secretagogue as it is the primary fuel source in food. Glucose metabolism is central to generation of MCFs that lead to insulin release, most notably ATP. In addition, other classes of nutrients are able to augment insulin secretion and these include members of the lipid and amino acid family of nutrients. Therefore, it is important to investigate the interplay between glucose, lipid, and amino acid metabolism, as it is this mixed nutrient sensing that generate the MCFs required for insulin exocytosis. The mechanisms by which these nutrients are metabolized to generate MCFs, and how they impact on β-cell insulin release and function, are discussed in detail in this article.
Collapse
Affiliation(s)
- Kevin Keane
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, Western Australia, Australia
| | - Philip Newsholme
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, Western Australia, Australia.
| |
Collapse
|
37
|
Zhou Y, Waanders LF, Holmseth S, Guo C, Berger UV, Li Y, Lehre AC, Lehre KP, Danbolt NC. Proteome analysis and conditional deletion of the EAAT2 glutamate transporter provide evidence against a role of EAAT2 in pancreatic insulin secretion in mice. J Biol Chem 2013; 289:1329-44. [PMID: 24280215 DOI: 10.1074/jbc.m113.529065] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Islet function is incompletely understood in part because key steps in glutamate handling remain undetermined. The glutamate (excitatory amino acid) transporter 2 (EAAT2; Slc1a2) has been hypothesized to (a) provide islet cells with glutamate, (b) protect islet cells against high extracellular glutamate concentrations, (c) mediate glutamate release, or (d) control the pH inside insulin secretory granules. Here we floxed the EAAT2 gene to produce the first conditional EAAT2 knock-out mice. Crossing with Nestin-cyclization recombinase (Cre) eliminated EAAT2 from the brain, resulting in epilepsy and premature death, confirming the importance of EAAT2 for brain function and validating the genetic construction. Crossing with insulin-Cre lines (RIP-Cre and IPF1-Cre) to obtain pancreas-selective deletion did not appear to affect survival, growth, glucose tolerance, or β-cell number. We found (using TaqMan RT-PCR, immunoblotting, immunocytochemistry, and proteome analysis) that the EAAT2 levels were too low to support any of the four hypothesized functions. The proteome analysis detected more than 7,000 islet proteins of which more than 100 were transporters. Although mitochondrial glutamate transporters and transporters for neutral amino acids were present at high levels, all other transporters with known ability to transport glutamate were strikingly absent. Glutamate-metabolizing enzymes were abundant. The level of glutamine synthetase was 2 orders of magnitude higher than that of glutaminase. Taken together this suggests that the uptake of glutamate by islets from the extracellular fluid is insignificant and that glutamate is intracellularly produced. Glutamine synthetase may be more important for islets than assumed previously.
Collapse
Affiliation(s)
- Yun Zhou
- From The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Nongonierma AB, Gaudel C, Murray BA, Flynn S, Kelly PM, Newsholme P, FitzGerald RJ. Insulinotropic properties of whey protein hydrolysates and impact of peptide fractionation on insulinotropic response. Int Dairy J 2013. [DOI: 10.1016/j.idairyj.2013.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
39
|
Gaudel C, Nongonierma AB, Maher S, Flynn S, Krause M, Murray BA, Kelly PM, Baird AW, FitzGerald RJ, Newsholme P. A whey protein hydrolysate promotes insulinotropic activity in a clonal pancreatic β-cell line and enhances glycemic function in ob/ob mice. J Nutr 2013; 143:1109-14. [PMID: 23658425 DOI: 10.3945/jn.113.174912] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Whey protein hydrolysates (WPHs) represent novel antidiabetic agents that affect glycemia in animals and humans, but little is known about their insulinotropic effects. The effects of a WPH were analyzed in vitro on acute glucose-induced insulin secretion in pancreatic BRIN-BD11 β cells. WPH permeability across Caco-2 cell monolayers was determined in a 2-tiered intestinal model. WPH effects on insulin resistance were studied in vivo following an 8-wk oral ingestion (100 mg/kg body weight) by ob/ob (OB-WPH) and wild-type mice (WT-WPH) compared with vehicle control (OB and WT groups) using a 2 × 2 factorial design, genotype × treatment. BRIN-BD11 cells showed a robust and reproducible dose-dependent insulinotropic effect of WPH (from 0.01 to 5.00 g/L). WPH bioactive constituents were permeable across Caco-2 cell monolayers. In the OB-WPH and WT-WPH groups, WPH administration improved glucose clearance after a glucose challenge (2 g/kg body weight), as indicated by differences in the area under curves (AUCs) (P ≤ 0.05). The basal plasma glucose concentration was not affected by WPH treatment in either genotype. The plasma insulin concentration was lower in the OB-WPH than in the OB group (P ≤ 0.005) but was similar between the WT and WT-WPH groups; the interaction genotype × treatment was significant (P ≤ 0.005). Insulin release from pancreatic islets isolated from the OB-WPH group was greater (P ≤ 0.005) than that from the OB group but did not differ between the WT-WPH and WT groups; the interaction genotype × treatment was not significant. In conclusion, an 8-wk oral administration of WPH improved blood glucose clearance, reduced hyperinsulinemia, and restored the pancreatic islet capacity to secrete insulin in response to glucose in ob/ob mice. Hence, it may be useful in diabetes management.
Collapse
Affiliation(s)
- Celine Gaudel
- School of Biomolecular and Biomedical Sciences, Health Sciences Centre, University College Dublin, Belfield, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Sailer M, Dahlhoff C, Giesbertz P, Eidens MK, de Wit N, Rubio-Aliaga I, Boekschoten MV, Müller M, Daniel H. Increased plasma citrulline in mice marks diet-induced obesity and may predict the development of the metabolic syndrome. PLoS One 2013; 8:e63950. [PMID: 23691124 PMCID: PMC3653803 DOI: 10.1371/journal.pone.0063950] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 04/10/2013] [Indexed: 12/31/2022] Open
Abstract
In humans, plasma amino acid concentrations of branched-chain amino acids (BCAA) and aromatic amino acids (AAA) increase in states of obesity, insulin resistance and diabetes. We here assessed whether these putative biomarkers can also be identified in two different obesity and diabetic mouse models. C57BL/6 mice with diet-induced obesity (DIO) mimic the metabolic impairments of obesity in humans characterized by hyperglycemia, hyperinsulinemia and hepatic triglyceride accumulation. Mice treated with streptozotocin (STZ) to induce insulin deficiency were used as a type 1 diabetes model. Plasma amino acid profiling of two high fat (HF) feeding trials revealed that citrulline and ornithine concentrations are elevated in obese mice, while systemic arginine bioavailability (ratio of plasma arginine to ornithine + citrulline) is reduced. In skeletal muscle, HF feeding induced a reduction of arginine levels while citrulline levels were elevated. However, arginine or citrulline remained unchanged in their key metabolic organs, intestine and kidney. Moreover, the intestinal conversion of labeled arginine to ornithine and citrulline in vitro remained unaffected by HF feeding excluding the intestine as prime site of these alterations. In liver, citrulline is mainly derived from ornithine in the urea cycle and DIO mice displayed reduced hepatic ornithine levels. Since both amino acids share an antiport mechanism for mitochondrial import and export, elevated plasma citrulline may indicate impaired hepatic amino acid handling in DIO mice. In the insulin deficient mice, plasma citrulline and ornithine levels also increased and additionally these animals displayed elevated BCAA and AAA levels like insulin resistant and diabetic patients. Therefore, type 1 diabetic mice but not DIO mice show the “diabetic fingerprint” of plasma amino acid changes observed in humans. Additionally, citrulline may serve as an early indicator of the obesity-dependent metabolic impairments.
Collapse
Affiliation(s)
- Manuela Sailer
- Molecular Nutrition Unit, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Christoph Dahlhoff
- Molecular Nutrition Unit, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- PhD Graduate School ‘Epigenetics, Imprinting and Nutrition’, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Pieter Giesbertz
- Molecular Nutrition Unit, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Mena K. Eidens
- Molecular Nutrition Unit, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Nicole de Wit
- Netherlands Nutrigenomics Centre, TI Food & Nutrition, Wageningen University, Wageningen, The Netherlands
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Isabel Rubio-Aliaga
- Molecular Nutrition Unit, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Mark V. Boekschoten
- Netherlands Nutrigenomics Centre, TI Food & Nutrition, Wageningen University, Wageningen, The Netherlands
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Michael Müller
- Netherlands Nutrigenomics Centre, TI Food & Nutrition, Wageningen University, Wageningen, The Netherlands
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Hannelore Daniel
- Molecular Nutrition Unit, Research Center for Nutrition and Food Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- * E-mail:
| |
Collapse
|
41
|
Salvucci M, Neufeld Z, Newsholme P. Mathematical model of metabolism and electrophysiology of amino acid and glucose stimulated insulin secretion: in vitro validation using a β-cell line. PLoS One 2013; 8:e52611. [PMID: 23520444 PMCID: PMC3592881 DOI: 10.1371/journal.pone.0052611] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 11/20/2012] [Indexed: 12/29/2022] Open
Abstract
We integrated biological experimental data with mathematical modelling to gain insights into the role played by L-alanine in amino acid-stimulated insulin secretion (AASIS) and in D-glucose-stimulated insulin secretion (GSIS), details important to the understanding of complex β-cell metabolic coupling relationships. We present an ordinary differential equations (ODEs) based simplified kinetic model of core metabolic processes leading to ATP production (glycolysis, TCA cycle, L-alanine-specific reactions, respiratory chain, ATPase and proton leak) and Ca(2+) handling (essential channels and pumps in the plasma membrane) in pancreatic β-cells and relate these to insulin secretion. Experimental work was performed using a clonal rat insulin-secreting cell line (BRIN-BD11) to measure the consumption or production of a range of important biochemical parameters (D-glucose, L-alanine, ATP, insulin secretion) and Ca(2+) levels. These measurements were then used to validate the theoretical model and fine-tune the parameters. Mathematical modelling was used to predict L-lactate and L-glutamate concentrations following D-glucose and/or L-alanine challenge and Ca(2+) levels upon stimulation with a non metabolizable L-alanine analogue. Experimental data and mathematical model simulations combined suggest that L-alanine produces a potent insulinotropic effect via both a stimulatory impact on β-cell metabolism and as a direct result of the membrane depolarization due to Ca(2+) influx triggered by L-alanine/Na(+) co-transport. Our simulations indicate that both high intracellular ATP and Ca(2+) concentrations are required in order to develop full insulin secretory responses. The model confirmed that K(+) ATP channel independent mechanisms of stimulation of intracellular Ca(2+) levels, via generation of mitochondrial coupling messengers, are essential for promotion of the full and sustained insulin secretion response in β-cells.
Collapse
Affiliation(s)
- Manuela Salvucci
- School of Biomolecular and Biomedical Sciences, University College Dublin, Dublin, Ireland.
| | | | | |
Collapse
|
42
|
Ranawana V, Kaur B. Role of proteins in insulin secretion and glycemic control. ADVANCES IN FOOD AND NUTRITION RESEARCH 2013; 70:1-47. [PMID: 23722093 DOI: 10.1016/b978-0-12-416555-7.00001-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dietary proteins are essential for the life of all animals and humans at all stages of the life cycle. They serve many structural and biochemical functions and have significant effects on health and wellbeing. Dietary protein consumption has shown an upward trend in developed countries in the past two decades primarily due to greater supply and affordability. Consumption is also on the rise in developing countries as affluence is increasing. Research shows that proteins have a notable impact on glucose homeostasis mechanisms, predominantly through their effects on insulin, incretins, gluconeogenesis, and gastric emptying. Since higher protein consumption and impaired glucose tolerance can be commonly seen in the same population demographics, a thorough understanding of the former's role in glucose homeostasis is crucial both toward the prevention and management of the latter. This chapter reviews the current state of the art on proteins, amino acids, and their effects on blood glucose and insulin secretion.
Collapse
Affiliation(s)
- Viren Ranawana
- Clinical Nutrition Research Centre, Singapore Institute for Clinical Sciences, Singapore, Singapore.
| | | |
Collapse
|
43
|
Amino acids potentiate insulin signaling in CHO-K1 at high glucose conditions. Arch Med Res 2012; 43:173-82. [PMID: 22609522 DOI: 10.1016/j.arcmed.2012.04.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 03/26/2012] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Amino acids reportedly increase the glucose uptake under high glucose conditions. However, there are controversies in the role of amino acids in diabetes mellitus. The present study explores the insulin signaling pathway involved in glucose uptake mediated by amino acids in CHO-K1 cells. METHODS CHO-K1 cells were exposed to normal (7 mM) and high glucose (17 and 27 mM) with 100 nM insulin in the presence and absence of amino acid mixtures (AAM) in varying concentration (5 and 20 mM) followed by the assays, insulin receptor tyrosine kinase (IRTK) and phosphatidylinositol 3 kinase (PI3K) by autoradiography, protein kinase B (Akt) and glucose transporter (GLUT4) by Western blot and glycogen synthase (GS) by HPLC. RESULTS The addition of 5 and 20 mM AAM significantly increased IRTK and PI3K activity (ANOVA p = 0.025, p = 0.003, respectively) with increasing glucose concentration. Addition of 5 mM AAM in the presence of normal glucose significantly increased the levels of phosphorylated Akt Ser473 (p = 0.02) with no significant change at high glucose. At 20 mM AAM there was a significant decrease in Akt phosphorylation (p = 0.035) that was increased by high glucose concentration. GLUT4 protein levels were increased with AAM (5 mM) along with increase in glycogen synthase activity at all glucose concentrations (p <0.05). CONCLUSIONS Amino acids as a mixture is beneficial in augmenting insulin signaling pathway via IRTK/PI3K/GLUT4 pathway along with activation of GS in CHO-K1 cells, thereby ensuring increased intracellular glucose availability.
Collapse
|
44
|
Mel M, Karim MIA, Yusuf SAM, Hashim YZHY, Ahmad Nor Y. Comparing BRIN-BD11 culture producing insulin using different type of microcarriers. Cytotechnology 2010; 62:423-30. [PMID: 20953703 DOI: 10.1007/s10616-010-9294-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Accepted: 07/27/2010] [Indexed: 11/29/2022] Open
Abstract
This research was conducted to examine the growth profile, growth kinetics, and insulin-secretory responsiveness of BRIN-BD11 cells grown in optimized medium on different types of microcarriers (MCs). Comparisons were made on modified polystyrene (Hillex(®) II) and crosslinked polystyrene Plastic Plus (PP) from Solohill Engineering. The cell line producing insulin was cultured in a 25 cm(2) T-flask as control while MCs based culture was implemented in a stirred tank bioreactor with 1 L working volume. For each culture type, the viable cell number, glucose, lactate, glutamate, and insulin concentrations were measured and compared. Maximum viable cell number was obtained at 1.47 × 10(5) cell/mL for PP microcarrier (PPMCs) culture, 1.35 × 10(5) cell/mL Hillex(®) II (HIIMCs) culture and 0.95 × 10(5) cell/mL for T-flask culture, respectively. The highest insulin concentration has been produced in PPMCs culture (5.31 mg/L) compared to HIIMCs culture (2.01 mg/L) and T-flask culture (1.99 mg/L). Therefore overall observation suggested that PPMCs was likely preferred to be used for BRIN-BD11 cell culture as compared with Hillex(®) II MCs.
Collapse
Affiliation(s)
- Maizirwan Mel
- Bioprocess and Molecular Engineering Research Unit, Department of Biotechnology Engineering, Faculty of Engineering, International Islamic University Malaysia, P.O. Box 10, 50728, Kuala Lumpur, Malaysia,
| | | | | | | | | |
Collapse
|
45
|
Amino acids influence the glucose uptake through GLUT4 in CHO-K1 cells under high glucose conditions. Mol Cell Biochem 2010; 344:43-53. [PMID: 20628794 DOI: 10.1007/s11010-010-0527-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 06/22/2010] [Indexed: 01/11/2023]
Abstract
According to studies earlier, amino acids have proven to be antidiabetic, antiglycating, and anticataractogenic. The present study was to explore whether amino acids as mixtures could enhance glucose uptake in CHO-K1 cells specifically. The cells in F-12K1 serum-free medium were exposed to normal (7 mM) and high glucose (12, 17 and 27 mM) in the presence and absence of amino acids mixture (AAM) in varying concentration (2.5, 5 and 10 mM). The mixture 5 and 10 mM AAM increased the 2-deoxyglucose (2DG) uptake at all glucose concentration significantly. There was also a significant increase in the GLUT4 (glucose transporter) translocation as revealed by flow cytometer. Addition of a mixture of amino acids was found to improve cell viability, which got altered by high glucose in the CHO-K1 cells. Amino acids as mixture had a beneficial effect in improving the net utilization of glucose as an additive effect with insulin.
Collapse
|
46
|
Nutrient regulation of insulin secretion and beta-cell functional integrity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:91-114. [PMID: 20217496 DOI: 10.1007/978-90-481-3271-3_6] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Pancreatic beta-cells are often referred to as "fuel sensors" as they continually monitor and respond to dietary nutrients, under the modulation of additional neurohormonal signals, in order to secrete insulin to best meet the needs of the organism. beta-cell nutrient sensing requires metabolic activation, resulting in production of stimulus-secretion coupling signals that promote insulin biosynthesis and release. The primary stimulus for insulin secretion is glucose, and islet beta-cells are particularly responsive to this important nutrient secretagogue, It is important to consider individual effects of different classes of nutrient or other physiological or pharmacological agents on metabolism and insulin secretion. However, given that beta-cells are continually exposed to a complex milieu of nutrients and other circulating factors, it is important to also acknowledge and examine the interplay between glucose metabolism and that of the two other primary nutrient classes, the amino acids and fatty acids. It is the mixed nutrient sensing and outputs of glucose, amino and fatty acid metabolism that generate the metabolic coupling factors (MCFs) involved in signaling for insulin exocytosis. Primary MCFs in the beta-cell include ATP, NADPH, glutamate, long chain acyl-CoA and diacylglycerol and are discussed in detail in this article.
Collapse
|
47
|
Overexpression of the malate-aspartate NADH shuttle member Aralar1 in the clonal beta-cell line BRIN-BD11 enhances amino-acid-stimulated insulin secretion and cell metabolism. Clin Sci (Lond) 2009; 117:321-30. [PMID: 19344310 PMCID: PMC2782311 DOI: 10.1042/cs20090126] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the present study, we have investigated the effects of the transduction with recombinant adenovirus AdCA-Aralar1 (aspartate–glutamate carrier 1) on the metabolism, function and secretory properties of the glucose- and amino-acid-responsive clonal insulin-secreting cell line BRIN-BD11. Aralar1 overexpression increased long-term (24 h) and acute (20 min) glucose- and amino-acid-stimulated insulin secretion, cellular glucose metabolism, L-alanine and L-glutamine consumption, cellular ATP and glutamate concentrations, and stimulated glutamate release. However, cellular triacylglycerol and glycogen contents were decreased as was lactate production. These findings indicate that increased malate–aspartate shuttle activity positively shifted β-cell metabolism, thereby increasing glycolysis capacity, stimulus–secretion coupling and, ultimately, enhancing insulin secretion. We conclude that Aralar1 is a key metabolic control site in insulin-secreting cells.
Collapse
|
48
|
Gammelsaeter R, Jenstad M, Bredahl MKL, Gundersen V, Chaudhry FA. Complementary expression of SN1 and SAT2 in the islets of Langerhans suggests concerted action of glutamine transport in the regulation of insulin secretion. Biochem Biophys Res Commun 2009; 381:378-82. [PMID: 19233140 DOI: 10.1016/j.bbrc.2009.02.062] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 02/11/2009] [Indexed: 11/29/2022]
Abstract
Insulin and glucagon secretion from the islets of Langerhans is highly regulated. Although an increased plasma glucose level is the major stimulus for insulin exocytosis, roles for glutamine and glutamate have been suggested. Interestingly, the islet cells display elements associated with synaptic transmission. In the central nervous system (CNS), glutamine transport by SN1 and SAT2 sustain the generation of neurotransmitter glutamate. We hypothesized that the same transporters are essential for glutamine transport into the islet cells and for subsequent formation of glutamate acting as an intracellular signaling molecule. We demonstrate that islet cells express several transporters which can mediate glutamine transport. In particular, we show pronounced expression of SN1 and SAT2 in B-cells and A-cells, respectively. The cell-specific expression of these transporters together with their functional characteristics suggest an important role for glutamine in the regulation of insulin secretion.
Collapse
Affiliation(s)
- R Gammelsaeter
- The Centre for Molecular Biology and Neuroscience, University of Oslo, N-0317 Oslo, Norway
| | | | | | | | | |
Collapse
|
49
|
Liu Z, Jeppesen PB, Gregersen S, Chen X, Hermansen K. Dose- and Glucose-Dependent Effects of Amino Acids on Insulin Secretion from Isolated Mouse Islets and Clonal INS-1E Beta-Cells. Rev Diabet Stud 2009; 5:232-44. [PMID: 19290384 DOI: 10.1900/rds.2008.5.232] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The influence of glucose and fatty acids on beta-cell function is well established whereas little is known about the role of amino acids (AAs). METHODS Islets isolated from NMRI mice were incubated overnight. After preincubation, isolated islets as well as clonal INS-1E beta-cells were incubated for 60 min in a modified Krebs Ringer buffer containing glucose and AAs. RESULTS At 16.7 mmol/l (mM) glucose, L-arginine, L-lysine, L-alanine, L-proline, L-leucine, and L-glutamine potentiated glucose-stimulated insulin secretion dose-dependently, while DL-homocysteine inhibited insulin secretion. Maximal insulin stimulation was obtained at 20 mM L-proline, L-lysine, L-alanine, L-arginine (islets: 2.5 to 6.7 fold increase; INS-1E cells: 1.6 to 2.2 fold increase). L-glutamine and L-leucine only increased glucose-stimulated (16.7 mM) insulin secretion (INS-1E cells: 1.5 and 1.3 fold, respectively) at an AA concentration of 20 mM. Homocysteine inhibited insulin secretion both at 5.6 mM and 16.7 mM glucose. At glucose levels ranging from 1.1 to 25 mM, the equimolar concentration of 10 mM, L-proline, L-lysine, L-arginine increased insulin secretion from mouse islets and INS-1E cells at all glucose levels applied, with a maximal effect obtained at 25 mM glucose. At a concentration of 10 mM, L-arginine and L-lysine had the highest insulinotropic potency among the AAs investigated. CONCLUSION L-arginine, L-lysine, L-alanine, L-proline, L-leucine and L-glutamine acutely stimulate insulin secretion from mouse islets and INS-1E cells in a dose- and glucose-dependent manner, whereas DL-homocysteine inhibits insulin release.
Collapse
Affiliation(s)
- Zhenping Liu
- Department of Endocrinology and Metabolism C, Aarhus University Hospital, Aarhus Sygehus THG, Tage-Hansens Gade 2, DK-8000 Aarhus C, Denmark
| | | | | | | | | |
Collapse
|
50
|
Prolonged L-alanine exposure induces changes in metabolism, Ca2+ handling and desensitization of insulin secretion in clonal pancreatic β-cells. Clin Sci (Lond) 2009; 116:341-51. [DOI: 10.1042/cs20080138] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute insulin-releasing actions of amino acids have been studied in detail, but comparatively little is known about the β-cell effects of long-term exposure to amino acids. The present study examined the effects of prolonged exposure of β-cells to the metabolizable amino acid L-alanine. Basal insulin release or cellular insulin content were not significantly altered by alanine culture, but acute alanine-induced insulin secretion was suppressed by 74% (P<0.001). Acute stimulation of insulin secretion with glucose, KCl or KIC (2-oxoisocaproic acid) following alanine culture was not affected. Acute alanine exposure evoked strong cellular depolarization after control culture, whereas AUC (area under the curve) analysis revealed significant (P<0.01) suppression of this action after culture with alanine. Compared with control cells, prior exposure to alanine also markedly decreased (P<0.01) the acute elevation of [Ca2+]i (intracellular [Ca2+]) induced by acute alanine exposure. These diminished stimulatory responses were partially restored after 18 h of culture in the absence of alanine, indicating reversible amino-acid-induced desensitization. 13C NMR spectra revealed that alanine culture increased glutamate labelling at position C4 (by 60%; P<0.01), as a result of an increase in the singlet peak, indicating increased flux through pyruvate dehydrogenase. Consistent with this, protein expression of the pyruvate dehydrogenase kinases PDK2 and PDK4 was significantly reduced. This was accompanied by a decrease in cellular ATP (P<0.05), consistent with diminished insulin-releasing actions of this amino acid. Collectively, these results illustrate the phenomenon of β-cell desensitization by amino acids, indicating that prolonged exposure to alanine can induce reversible alterations to metabolic flux, Ca2+ handling and insulin secretion.
Collapse
|