1
|
Zhu X, Chen C, Liu Q, Zhu Z, Wu X, Zhang Y. Multiple pesticide exposure and impaired glucose regulation in U.S. non-diabetic population. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125519. [PMID: 39672370 DOI: 10.1016/j.envpol.2024.125519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
Prediabetes is a serious metabolic disorder that is often overlooked and 70% of individuals with prediabetes would eventually develop type 2 diabetes. The diabetogenic effects of pesticides have been reported in toxicological studies but their association with prediabetes is rarely investigated. We aimed to evaluate the association between pesticide exposure and impaired glucose regulation (IGR), including prediabetes (defined as impaired fasting glucose [IFG] and/or impaired glucose tolerance [IGT]) and insulin resistance, in a general U.S. non-diabetic population. Three classes of urinary pesticides, including organophosphorus pesticides (OPs), pyrethroid, and herbicides were measured. Generalized linear regression, restricted cubic spline, and Bayesian kernel machine regression (BKMR) models were combined to evaluate their associations. 3,5,6-trichloropyridinol (TCPY) was positively associated with prediabetes and IGT (highest vs lowest TCPY quartile: prediabetes: OR: 1.97, 95% CI: 1.18, 3.31; IGT: OR: 2.03, 95% CI: 1.14, 3.66) in a linear dose-response manner (P for nonlinear<0.05). Another two metabolites of OPs, malathion dicarboxylic acid (MDCA) diacid and para-nitrophenol (PNP), were found to increase the odds ratio of insulin resistance (PNP: OR: 1.22, 95% CI: 1.05, 1.42; MDCA: OR: 1.36, 95% CI: 1.08, 1.70) with linear dose-response curves (P for nonlinear<0.05). Considering mutual exposure to multiple pesticides, TCPY, MDCA, and PNP made the most contributions in the mixture exposure and IGR. No obvious interactions among pesticides were found in the multiple exposure settings. The odds ratio of TCPY exposure and prediabetes was increased with advancing age but not related to body mass index (BMI). The results remained robust in sensitivity analysis with restricted participants without abnormal urinary creatinine and unsteady glucose or insulin levels. Our findings suggested the close relationship between OPs and impaired glucose regulation, especially in older adults, which provides insights into the prevention of diabetes at the earlier stage.
Collapse
Affiliation(s)
- Xingdi Zhu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China; First School of Clinical Medicine, Nanjing Medical University, Nanjing, 211100, China
| | - Congxin Chen
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Qi Liu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Zhihong Zhu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Xiaoli Wu
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China
| | - Yuqing Zhang
- Department of Obstetrics and Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| |
Collapse
|
2
|
Hsiao HY, Nien CY, Shiu RF, Chin WC, Yen TH. Microplastic and nanoplastic exposure and risk of diabetes mellitus. World J Clin Cases 2025; 13:98110. [PMID: 39866647 PMCID: PMC11577526 DOI: 10.12998/wjcc.v13.i3.98110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/28/2024] [Accepted: 10/21/2024] [Indexed: 11/12/2024] Open
Abstract
The issue of plastic pollutants has become a growing concern. Both microplastics (MPs) (particle size < 5 mm) and nanoplastics (NPs) (particle size < 1 µm) can cause DNA damage, cytotoxicity, and oxidative stress in various organisms. The primary known impacts of microplastic/nanoplastic are observed in the liver and respiratory system, leading to hepatotoxicity and chronic obstructive pulmonary disease. Although research on the effects of MPs and NPs on diabetes is still in its early stages, there are potential concerns. This editorial highlights the risk to diabetics from co-exposure to contaminants and MPs/NPs, supported by evidence from animal studies and the various chemical compositions of MPs/NPs.
Collapse
Affiliation(s)
- Hui-Yi Hsiao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
| | - Chung-Yi Nien
- Department of Life Sciences, National Central University, Zhongli 320317, Taoyuan, Taiwan
| | - Ruei-Feng Shiu
- Center of Excellence for The Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
- Institute of Marine Environment and Ecology, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Wei-Chun Chin
- Department of Chemical and Materials Engineering, University of California Merced, Merced, CA 95343, United States
| | - Tzung-Hai Yen
- Department of Nephrology, Clinical Poison Center, Chang Gung Memorial Hospital, Linkou 33305, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
3
|
Hamilton A, Zhang Q, Gao R, Hill TG, Salehi A, Knudsen JG, Draper MB, Johnson PRV, Rorsman P, Tarasov AI. Nicotinic Signaling Stimulates Glucagon Secretion in Mouse and Human Pancreatic α-Cells. Diabetes 2025; 74:53-64. [PMID: 39475504 DOI: 10.2337/db23-0809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/10/2024] [Indexed: 12/22/2024]
Abstract
Smoking is widely regarded as a risk factor for type 2 diabetes because nicotine contributes to insulin resistance by desensitizing the insulin receptors in muscle, liver, or fat. Little is known, however, about the immediate regulation of islet hormonal output by nicotine, an agonist of ionotropic cholinergic receptors. We investigated this by imaging cytosolic Ca2+ dynamics in mouse and human islets using confocal microscopy and measuring glucagon secretion in response to the alkaloid from isolated mouse islets. Nicotine acutely stimulated cytosolic Ca2+ in glucagon-secreting α-cells but not in insulin-secreting β-cells. The 2.8- ± 0.5-fold (P < 0.05) increase in Ca2+, observed in >70% of α-cells, correlated well with a 2.5- ± 0.3-fold stimulation of glucagon secretion. Nicotine-induced elevation of cytosolic Ca2+ relied on influx from the extracellular compartment rather than release of the cation from intracellular depots. Metabotropic cholinergic signaling, monitored at the level of intracellular diacylglycerol, was limited to 69% of α-cells versus 94% of β-cells. We conclude that parasympathetic regulation of pancreatic islet hormone release uses different signaling pathways in β-cells (metabotropic) and α-cells (metabotropic and ionotropic), resulting in the fine-tuning of acetylcholine-induced glucagon exocytosis. Sustained nicotinic stimulation is, therefore, likely to attenuate insulin sensitivity by increasing glucagon release. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Alexander Hamilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- Unit of Molecular Metabolism, Clinical Research Centre, Lund University Diabetes Centre, Lund University and Malmö University Hospital, Malmö, Sweden
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rui Gao
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Thomas G Hill
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
| | - Albert Salehi
- Unit of Molecular Metabolism, Clinical Research Centre, Lund University Diabetes Centre, Lund University and Malmö University Hospital, Malmö, Sweden
- Metabolic Research Unit, Department of Physiology, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Jakob G Knudsen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Matthew B Draper
- School of Biomedical Sciences, Ulster University, Coleraine, U.K
| | - Paul R V Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- Oxford Biomedical Research Centre, National Institute for Health Research, Oxford, U.K
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- Metabolic Research Unit, Department of Physiology, Institute of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
- School of Biomedical Sciences, Ulster University, Coleraine, U.K
- Oxford Biomedical Research Centre, National Institute for Health Research, Oxford, U.K
| | - Andrei I Tarasov
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford and Churchill Hospital, Oxford, U.K
- School of Biomedical Sciences, Ulster University, Coleraine, U.K
| |
Collapse
|
4
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
5
|
Kawana Y, Imai J, Morizawa YM, Ikoma Y, Kohata M, Komamura H, Sato T, Izumi T, Yamamoto J, Endo A, Sugawara H, Kubo H, Hosaka S, Munakata Y, Asai Y, Kodama S, Takahashi K, Kaneko K, Sawada S, Yamada T, Ito A, Niizuma K, Tominaga T, Yamanaka A, Matsui K, Katagiri H. Optogenetic stimulation of vagal nerves for enhanced glucose-stimulated insulin secretion and β cell proliferation. Nat Biomed Eng 2024; 8:808-822. [PMID: 37945752 PMCID: PMC11310082 DOI: 10.1038/s41551-023-01113-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 09/26/2023] [Indexed: 11/12/2023]
Abstract
The enhancement of insulin secretion and of the proliferation of pancreatic β cells are promising therapeutic options for diabetes. Signals from the vagal nerve regulate both processes, yet the effectiveness of stimulating the nerve is unclear, owing to a lack of techniques for doing it so selectively and prolongedly. Here we report two optogenetic methods for vagal-nerve stimulation that led to enhanced glucose-stimulated insulin secretion and to β cell proliferation in mice expressing choline acetyltransferase-channelrhodopsin 2. One method involves subdiaphragmatic implantation of an optical fibre for the photostimulation of cholinergic neurons expressing a blue-light-sensitive opsin. The other method, which suppressed streptozotocin-induced hyperglycaemia in the mice, involves the selective activation of vagal fibres by placing blue-light-emitting lanthanide microparticles in the pancreatic ducts of opsin-expressing mice, followed by near-infrared illumination. The two methods show that signals from the vagal nerve, especially from nerve fibres innervating the pancreas, are sufficient to regulate insulin secretion and β cell proliferation.
Collapse
Affiliation(s)
- Yohei Kawana
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Yosuke M Morizawa
- Super-network Brain Physiology, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Yoko Ikoma
- Super-network Brain Physiology, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Masato Kohata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Komamura
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toshihiro Sato
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohito Izumi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junpei Yamamoto
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Endo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroto Sugawara
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haremaru Kubo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Yuichiro Munakata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akira Ito
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kuniyasu Niizuma
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Neurosurgical Engineering and Translational Neuroscience, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihiro Yamanaka
- Department of Neuroscience II, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Ko Matsui
- Super-network Brain Physiology, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
6
|
Wang F, Yu J, Lin L, Lin D, Chen K, Quan H. A genome-wide association study identifies 25(OH)D3-associated genetic variants in the prediabetic Chinese population. Endocrine 2024; 84:1154-1163. [PMID: 38291318 DOI: 10.1007/s12020-024-03694-7] [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: 09/14/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
OBJECTIVES Diabetes mellitus has been a significant public health problem, associated with high rates of morbidity, disability, and mortality. Prediabetes is a crucial period for preventing and managing diabetes. 25(OH)D3 is an important risk factor for prediabetes. However, there is limited genetic knowledge of 25(OH)D3 in the Chinese population. This study was designed to identify genetic variants associated with 25(OH)D3 and explore the potential pathogenesis of prediabetes. METHODS In this study, 451 individuals with prediabetes were recruited to determine the genetic variants associated with 25(OH)D3 through a genome-wide association study (GWAS). Gene mapping and overrepresentation analysis (ORA) were further performed to explore the candidate genes and their biological mechanisms. RESULTS In this study, we identified two independent significant loci (rs9457733 and rs11243373, p < 5 × 10-6 and r2 < 0.6) and 37 candidate genes associated with 25(OH)D3 in prediabetes. Furthermore, the ORA analysis revealed that two genes in the gene sets, SLC22A1 and SLC22A3, were found to be significantly enriched in monoamine transmembrane transporter activity and quaternary ammonium group transmembrane transporter activity, as determined by WebGestalt and g:Profiler (padj < 0.05). CONCLUSION The identification of potential genes associated with 25(OH)D3 provides a foundation for a better understanding of the pathogenesis, diagnosis, and treatment of prediabetes.
Collapse
Affiliation(s)
- Fei Wang
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jingwen Yu
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Leweihua Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Danhong Lin
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Huibiao Quan
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China.
| |
Collapse
|
7
|
Hill TG, Hill DJ. The Importance of Intra-Islet Communication in the Function and Plasticity of the Islets of Langerhans during Health and Diabetes. Int J Mol Sci 2024; 25:4070. [PMID: 38612880 PMCID: PMC11012451 DOI: 10.3390/ijms25074070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Islets of Langerhans are anatomically dispersed within the pancreas and exhibit regulatory coordination between islets in response to nutritional and inflammatory stimuli. However, within individual islets, there is also multi-faceted coordination of function between individual beta-cells, and between beta-cells and other endocrine and vascular cell types. This is mediated partly through circulatory feedback of the major secreted hormones, insulin and glucagon, but also by autocrine and paracrine actions within the islet by a range of other secreted products, including somatostatin, urocortin 3, serotonin, glucagon-like peptide-1, acetylcholine, and ghrelin. Their availability can be modulated within the islet by pericyte-mediated regulation of microvascular blood flow. Within the islet, both endocrine progenitor cells and the ability of endocrine cells to trans-differentiate between phenotypes can alter endocrine cell mass to adapt to changed metabolic circumstances, regulated by the within-islet trophic environment. Optimal islet function is precariously balanced due to the high metabolic rate required by beta-cells to synthesize and secrete insulin, and they are susceptible to oxidative and endoplasmic reticular stress in the face of high metabolic demand. Resulting changes in paracrine dynamics within the islets can contribute to the emergence of Types 1, 2 and gestational diabetes.
Collapse
Affiliation(s)
- Thomas G. Hill
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - David J. Hill
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada;
- Departments of Medicine, Physiology and Pharmacology, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
8
|
Salau VF, Erukainure OL, Olofinsan KO, Msomi NZ, Ijomone OM, Islam MS. Vanillin improves glucose homeostasis and modulates metabolic activities linked to type 2 diabetes in fructose-streptozotocin induced diabetic rats. Arch Physiol Biochem 2024; 130:169-182. [PMID: 34752171 DOI: 10.1080/13813455.2021.1988981] [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: 08/03/2021] [Accepted: 09/29/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE This study investigated the antidiabetic effect of vanillin using in vitro, in silico, and in vivo experimental models. METHODOLOGY Type 2 diabetes (T2D) was induced in male Sprague-Dawley (SD) rats using fructose-streptozotocin (STZ), then orally administered low (150 mg/kg bodyweight) or high (300 mg/kg bodyweight) dose of vanillin for 5 weeks intervention period. RESULTS Vanillin suppressed the levels of blood glucose, serum cholesterol, triglyceride, low-density lipoprotein cholesterol (LDL-c), alanine transaminase (ALT), aspartate transaminase (AST), creatinine, urea, uric acid, when elevated serum insulin, HDL-cholesterol, and concomitantly improved pancreatic β-cell function, glucose tolerance, and pancreatic morphology. It also elevated both serum and pancreatic tissue GSH level, SOD and catalase activities, and hepatic glycogen level, while depleting malondialdehyde level, α-amylase, lipase, acetylcholinesterase, ATPase, ENTPDase and 5'-nucleotidase, glucose-6-phosphatase, fructose-1,6-bisphosphatase, and glycogen phosphorylase activities. CONCLUSIONS The results indicate the potent antidiabetic effect of vanillin against T2D and its associated complications.
Collapse
Affiliation(s)
- Veronica F Salau
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Biochemistry, Veritas University, Abuja, Nigeria
| | - Ochuko L Erukainure
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
- Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Kolawole O Olofinsan
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nontokozo Z Msomi
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| | | | - Md Shahidul Islam
- Department of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
9
|
Varney MJ, Benovic JL. The Role of G Protein-Coupled Receptors and Receptor Kinases in Pancreatic β-Cell Function and Diabetes. Pharmacol Rev 2024; 76:267-299. [PMID: 38351071 PMCID: PMC10877731 DOI: 10.1124/pharmrev.123.001015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 02/16/2024] Open
Abstract
Type 2 diabetes (T2D) mellitus has emerged as a major global health concern that has accelerated in recent years due to poor diet and lifestyle. Afflicted individuals have high blood glucose levels that stem from the inability of the pancreas to make enough insulin to meet demand. Although medication can help to maintain normal blood glucose levels in individuals with chronic disease, many of these medicines are outdated, have severe side effects, and often become less efficacious over time, necessitating the need for insulin therapy. G protein-coupled receptors (GPCRs) regulate many physiologic processes, including blood glucose levels. In pancreatic β cells, GPCRs regulate β-cell growth, apoptosis, and insulin secretion, which are all critical in maintaining sufficient β-cell mass and insulin output to ensure euglycemia. In recent years, new insights into the signaling of incretin receptors and other GPCRs have underscored the potential of these receptors as desirable targets in the treatment of diabetes. The signaling of these receptors is modulated by GPCR kinases (GRKs) that phosphorylate agonist-activated GPCRs, marking the receptor for arrestin binding and internalization. Interestingly, genome-wide association studies using diabetic patient cohorts link the GRKs and arrestins with T2D. Moreover, recent reports show that GRKs and arrestins expressed in the β cell serve a critical role in the regulation of β-cell function, including β-cell growth and insulin secretion in both GPCR-dependent and -independent pathways. In this review, we describe recent insights into GPCR signaling and the importance of GRK function in modulating β-cell physiology. SIGNIFICANCE STATEMENT: Pancreatic β cells contain a diverse array of G protein-coupled receptors (GPCRs) that have been shown to improve β-cell function and survival, yet only a handful have been successfully targeted in the treatment of diabetes. This review discusses recent advances in our understanding of β-cell GPCR pharmacology and regulation by GPCR kinases while also highlighting the necessity of investigating islet-enriched GPCRs that have largely been unexplored to unveil novel treatment strategies.
Collapse
Affiliation(s)
- Matthew J Varney
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Röthe J, Kraft R, Ricken A, Kaczmarek I, Matz-Soja M, Winter K, Dietzsch AN, Buchold J, Ludwig MG, Liebscher I, Schöneberg T, Thor D. The adhesion GPCR GPR116/ADGRF5 has a dual function in pancreatic islets regulating somatostatin release and islet development. Commun Biol 2024; 7:104. [PMID: 38228886 PMCID: PMC10791652 DOI: 10.1038/s42003-024-05783-9] [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: 11/03/2023] [Accepted: 01/05/2024] [Indexed: 01/18/2024] Open
Abstract
Glucose homeostasis is maintained by hormones secreted from different cell types of the pancreatic islets and controlled by manifold input including signals mediated through G protein-coupled receptors (GPCRs). RNA-seq analyses revealed expression of numerous GPCRs in mouse and human pancreatic islets, among them Gpr116/Adgrf5. GPR116 is an adhesion GPCR mainly found in lung and required for surfactant secretion. Here, we demonstrate that GPR116 is involved in the somatostatin release from pancreatic delta cells using a whole-body as well as a cell-specific knock-out mouse model. Interestingly, the whole-body GPR116 deficiency causes further changes such as decreased beta-cell mass, lower number of small islets, and reduced pancreatic insulin content. Glucose homeostasis in global GPR116-deficient mice is maintained by counter-acting mechanisms modulating insulin degradation. Our data highlight an important function of GPR116 in controlling glucose homeostasis.
Collapse
Affiliation(s)
- Juliane Röthe
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Robert Kraft
- Carl-Ludwig-Institute for Physiology, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Albert Ricken
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Isabell Kaczmarek
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Madlen Matz-Soja
- Medical Department II - Gastroenterology, Hepatology, Infectious Diseases, Pneumology, University Medical Center, Leipzig, Germany
- Division of Hepatology, Clinic and Polyclinic for Oncology, Gastroenterology, Hepatology, Infectious Diseases, and Pneumology, University Hospital, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Medical Faculty, Leipzig University, Leipzig, Germany
| | - André Nguyen Dietzsch
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Julia Buchold
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | | | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany.
| |
Collapse
|
11
|
Cazzola M, Rogliani P, Ora J, Calzetta L, Lauro D, Matera MG. Hyperglycaemia and Chronic Obstructive Pulmonary Disease. Diagnostics (Basel) 2023; 13:3362. [PMID: 37958258 PMCID: PMC10650064 DOI: 10.3390/diagnostics13213362] [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: 09/25/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) may coexist with type 2 diabetes mellitus (T2DM). Patients with COPD have an increased risk of developing T2DM compared with a control but, on the other side, hyperglycaemia and DM have been associated with reduced predicted levels of lung function. The mechanistic relationships between these two diseases are complicated, multifaceted, and little understood, yet they can impact treatment strategy. The potential risks and benefits for patients with T2DM treated with pulmonary drugs and the potential pulmonary risks and benefits for patients with COPD when taking antidiabetic drugs should always be considered. The interaction between the presence and/or treatment of COPD, risk of infection, presence and/or treatment of T2DM and risk of acute exacerbations of COPD (AECOPDs) can be represented as a vicious circle; however, several strategies may help to break this circle. The most effective approach to simultaneously treating T2DM and COPD is to interfere with the shared inflammatory substrate, thus targeting both lung inflammation (COPD) and vascular inflammation (DM). In any case, it is always crucial to establish glycaemic management since the reduction in lung function found in people with diabetes might decrease the threshold for clinical manifestations of COPD. In this article, we examine possible connections between COPD and T2DM as well as pharmacological strategies that could focus on these connections.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy
- Division of Respiratory Medicine, University Hospital Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Josuel Ora
- Division of Respiratory Medicine, University Hospital Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Disease and Lung Function, Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Davide Lauro
- Unit of Endocrinology and Metabolic Diseases, Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00173 Rome, Italy
- Division of Endocrinology and Diabetes, University Hospital Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania ‘Luigi Vanvitelli’, 81138 Naples, Italy
| |
Collapse
|
12
|
Chen Z, Liu XA, Kenny PJ. Central and peripheral actions of nicotine that influence blood glucose homeostasis and the development of diabetes. Pharmacol Res 2023; 194:106860. [PMID: 37482325 DOI: 10.1016/j.phrs.2023.106860] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cigarette smoking has long been recognized as a risk factor for type 2 diabetes (T2D), although the precise causal mechanisms underlying this relationship remain poorly understood. Recent evidence suggests that nicotine, the primary reinforcing component in tobacco, may play a pivotal role in connecting cigarette smoking and T2D. Extensive research conducted in both humans and animals has demonstrated that nicotine can elevate blood glucose levels, disrupt glucose homeostasis, and induce insulin resistance. The review aims to elucidate the genetic variants of nicotinic acetylcholine receptors associated with diabetes risk and provide a comprehensive overview of the available data on the mechanisms through which nicotine influences blood glucose homeostasis and the development of diabetes. Here we emphasize the central and peripheral actions of nicotine on the release of glucoregulatory hormones, as well as its effects on glucose tolerance and insulin sensitivity. Notably, the central actions of nicotine within the brain, which encompass both insulin-dependent and independent mechanisms, are highlighted as potential targets for intervention strategies in diabetes management.
Collapse
Affiliation(s)
- Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xin-An Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; University of Chinese Academy of Sciences, Beijing, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
13
|
Pettway YD, Saunders DC, Brissova M. The human α cell in health and disease. J Endocrinol 2023; 258:e220298. [PMID: 37114672 PMCID: PMC10428003 DOI: 10.1530/joe-22-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/27/2023] [Indexed: 04/29/2023]
Abstract
In commemoration of 100 years since the discovery of glucagon, we review current knowledge about the human α cell. Alpha cells make up 30-40% of human islet endocrine cells and play a major role in regulating whole-body glucose homeostasis, largely through the direct actions of their main secretory product - glucagon - on peripheral organs. Additionally, glucagon and other secretory products of α cells, namely acetylcholine, glutamate, and glucagon-like peptide-1, have been shown to play an indirect role in the modulation of glucose homeostasis through autocrine and paracrine interactions within the islet. Studies of glucagon's role as a counterregulatory hormone have revealed additional important functions of the α cell, including the regulation of multiple aspects of energy metabolism outside that of glucose. At the molecular level, human α cells are defined by the expression of conserved islet-enriched transcription factors and various enriched signature genes, many of which have currently unknown cellular functions. Despite these common threads, notable heterogeneity exists amongst human α cell gene expression and function. Even greater differences are noted at the inter-species level, underscoring the importance of further study of α cell physiology in the human context. Finally, studies on α cell morphology and function in type 1 and type 2 diabetes, as well as other forms of metabolic stress, reveal a key contribution of α cell dysfunction to dysregulated glucose homeostasis in disease pathogenesis, making targeting the α cell an important focus for improving treatment.
Collapse
Affiliation(s)
- Yasminye D. Pettway
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, 37232, USA
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, 37232, USA
| |
Collapse
|
14
|
Aldous N, Moin ASM, Abdelalim EM. Pancreatic β-cell heterogeneity in adult human islets and stem cell-derived islets. Cell Mol Life Sci 2023; 80:176. [PMID: 37270452 DOI: 10.1007/s00018-023-04815-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/27/2023] [Accepted: 05/19/2023] [Indexed: 06/05/2023]
Abstract
Recent studies reported that pancreatic β-cells are heterogeneous in terms of their transcriptional profiles and their abilities for insulin secretion. Sub-populations of pancreatic β-cells have been identified based on the functionality and expression of specific surface markers. Under diabetes condition, β-cell identity is altered leading to different β-cell sub-populations. Furthermore, cell-cell contact between β-cells and other endocrine cells within the islet play an important role in regulating insulin secretion. This highlights the significance of generating a cell product derived from stem cells containing β-cells along with other major islet cells for treating patients with diabetes, instead of transplanting a purified population of β-cells. Another key question is how close in terms of heterogeneity are the islet cells derived from stem cells? In this review, we summarize the heterogeneity in islet cells of the adult pancreas and those generated from stem cells. In addition, we highlight the significance of this heterogeneity in health and disease conditions and how this can be used to design a stem cell-derived product for diabetes cell therapy.
Collapse
Affiliation(s)
- Noura Aldous
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, Doha, Qatar
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, PO Box 34110, Doha, Qatar
| | - Abu Saleh Md Moin
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, PO Box 34110, Doha, Qatar
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, Kingdom of Bahrain
| | - Essam M Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, Doha, Qatar.
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Education City, PO Box 34110, Doha, Qatar.
| |
Collapse
|
15
|
Wei Y, Wang L, Liu J. The diabetogenic effects of pesticides: Evidence based on epidemiological and toxicological studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023:121927. [PMID: 37268216 DOI: 10.1016/j.envpol.2023.121927] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
While the use of pesticides has improved grain productivity and controlled vector-borne diseases, the widespread use of pesticides has resulted in ubiquitous environmental residues that pose health risks to humans. A number of studies have linked pesticide exposure to diabetes and glucose dyshomeostasis. This article reviews the occurrence of pesticides in the environment and human exposure, the associations between pesticide exposures and diabetes based on epidemiological investigations, as well as the diabetogenic effects of pesticides based on the data from in vivo and in vitro studies. The potential mechanisms by which pesticides disrupt glucose homeostasis include induction of lipotoxicity, oxidative stress, inflammation, acetylcholine accumulation, and gut microbiota dysbiosis. The gaps between laboratory toxicology research and epidemiological studies lead to an urgent research need on the diabetogenic effects of herbicides and current-use insecticides, low-dose pesticide exposure research, the diabetogenic effects of pesticides in children, and assessment of toxicity and risks of combined exposure to multiple pesticides with other chemicals.
Collapse
Affiliation(s)
- Yile Wei
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Linping Wang
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jing Liu
- MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
16
|
Fu Q, Jiang H, Qian Y, Lv H, Dai H, Zhou Y, Chen Y, He Y, Gao R, Zheng S, Liang Y, Li S, Xu X, Xu K, Yang T. Single-cell RNA sequencing combined with single-cell proteomics identifies the metabolic adaptation of islet cell subpopulations to high-fat diet in mice. Diabetologia 2023; 66:724-740. [PMID: 36538064 PMCID: PMC9765371 DOI: 10.1007/s00125-022-05849-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022]
Abstract
AIMS/HYPOTHESIS Islets have complex heterogeneity and subpopulations. Cell surface markers representing alpha, beta and delta cell subpopulations are urgently needed for investigations to explore the compositional changes of each subpopulation in obesity progress and diabetes onset, and the adaptation mechanism of islet metabolism induced by a high-fat diet (HFD). METHODS Single-cell RNA sequencing (scRNA-seq) was applied to identify alpha, beta and delta cell subpopulation markers in an HFD-induced mouse model of glucose intolerance. Flow cytometry and immunostaining were used to sort and assess the proportion of each subpopulation. Single-cell proteomics was performed on sorted cells, and the functional status of each alpha, beta and delta cell subpopulation in glucose intolerance was deeply elucidated based on protein expression. RESULTS A total of 33,999 cells were analysed by scRNA-seq and clustered into eight populations, including alpha, beta and delta cells. For alpha cells, scRNA-seq revealed that the Ace2low subpopulation had downregulated expression of genes related to alpha cell function and upregulated expression of genes associated with beta cell characteristics in comparison with the Ace2high subpopulation. The impaired function and increased fragility of ACE2low alpha cells exposure to HFD was further suggested by single-cell proteomics. As for beta cells, the CD81high subpopulation may indicate an immature signature of beta cells compared with the CD81low subpopulation, which had robust function. We also found differential expression of Slc2a2 in delta cells and a potentially stronger cellular function and metabolism in GLUT2low delta cells than GLUT2high delta cells. Moreover, an increased proportion of ACE2low alpha cells and CD81low beta cells, with a constant proportion of GLUT2low delta cells, were observed in HFD-induced glucose intolerance. CONCLUSIONS/INTERPRETATION We identified ACE2, CD81 and GLUT2 as surface markers to distinguish, respectively, alpha, beta and delta cell subpopulations with heterogeneous maturation and function. The changes in the proportion and functional status of islet endocrine subpopulations reflect the metabolic adaptation of islets to high-fat stress, which weakened the function of alpha cells and enhanced the function of beta and delta cells to bring about glycaemic homeostasis. Our findings provide a fundamental resource for exploring the mechanisms maintaining each islet endocrine subpopulation's fate and function in health and disease. DATA AVAILABILITY The scRNA-seq analysis datasets from the current study are available in the Gene Expression Omnibus (GEO) repository under the accession number GSE203376.
Collapse
Affiliation(s)
- Qi Fu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hemin Jiang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Qian
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Lv
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Dai
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuncai Zhou
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Chen
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunqiang He
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Gao
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuai Zheng
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yucheng Liang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Siqi Li
- BGI-Shenzhen, Shenzhen, China
- BGI-Wuhan Clinical Laboratories, BGI-Shenzhen, Wuhan, China
| | - Xinyu Xu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kuanfeng Xu
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Tao Yang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
17
|
Zhao Y, Veysman B. Revisiting the Pathogenesis of Type 1 Diabetes: Importance of Neural Input to Pancreatic Islets and the Therapeutic Capability of Stem Cell Educator TM Therapy to Restore Their Integrity. Biomedicines 2023; 11:594. [PMID: 36831130 PMCID: PMC9952924 DOI: 10.3390/biomedicines11020594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease with a shortage of islet β cells. To date, the etiology of T1D remains elusive. Increasing clinical evidence and animal studies demonstrate that autoimmune cells are directed against the nervous system of pancreatic islets, contributing to the development of T1D. Therefore, it highlights the necessity to explore novel clinical approaches to fundamentally correct the T1D autoimmunity not only focusing on islet β cells but also on protecting the islet nervous system. This allows the restoration of the integrity of islet innervation and the normal islet β-cell function. To address these issues, we developed a novel technology designated the Stem Cell Educator TM therapy, based on immune education by human cord-blood-derived multipotent stem cells (CB-SC). International amulticenter clinical trials demonstrated its clinical safety and efficacy to treat T1D and other autoimmune diseases. Stem Cell Educator TM therapy may have the potential to revolutionize the treatment of T1D, without the safety and ethical concerns associated with conventional immune and/or stem cell-based therapies.
Collapse
Affiliation(s)
- Yong Zhao
- Throne Biotechnologies, Paramus, NJ 07652, USA
| | | |
Collapse
|
18
|
Harvey KE, Tang S, LaVigne EK, Pratt EPS, Hockerman GH. RyR2 regulates store-operated Ca2+ entry, phospholipase C activity, and electrical excitability in the insulinoma cell line INS-1. PLoS One 2023; 18:e0285316. [PMID: 37141277 PMCID: PMC10159205 DOI: 10.1371/journal.pone.0285316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/19/2023] [Indexed: 05/05/2023] Open
Abstract
The ER Ca2+ channel ryanodine receptor 2 (RyR2) is required for maintenance of insulin content and glucose-stimulated insulin secretion, in part, via regulation of the protein IRBIT in the insulinoma cell line INS-1. Here, we examined store-operated and depolarization-dependent Ca2+entry using INS-1 cells in which either RyR2 or IRBIT were deleted. Store-operated Ca2+ entry (SOCE) stimulated with thapsigargin was reduced in RyR2KO cells compared to controls, but was unchanged in IRBITKO cells. STIM1 protein levels were not different between the three cell lines. Basal and stimulated (500 μM carbachol) phospholipase C (PLC) activity was also reduced specifically in RyR2KO cells. Insulin secretion stimulated by tolbutamide was reduced in RyR2KO and IRBITKO cells compared to controls, but was potentiated by an EPAC-selective cAMP analog in all three cell lines. Cellular PIP2 levels were increased and cortical f-actin levels were reduced in RyR2KO cells compared to controls. Whole-cell Cav channel current density was increased in RyR2KO cells compared to controls, and barium current was reduced by acute activation of the lipid phosphatase pseudojanin preferentially in RyR2KO cells over control INS-1 cells. Action potentials stimulated by 18 mM glucose were more frequent in RyR2KO cells compared to controls, and insensitive to the SK channel inhibitor apamin. Taken together, these results suggest that RyR2 plays a critical role in regulating PLC activity and PIP2 levels via regulation of SOCE. RyR2 also regulates β-cell electrical activity by controlling Cav current density and SK channel activation.
Collapse
Affiliation(s)
- Kyle E Harvey
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| | - Shiqi Tang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| | - Emily K LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, Indiana, United States of America
| | - Evan P S Pratt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Interdisciplinary Life Sciences Program, Purdue University, West Lafayette, Indiana, United States of America
| | - Gregory H Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| |
Collapse
|
19
|
Capozzi ME, D'Alessio DA, Campbell JE. The past, present, and future physiology and pharmacology of glucagon. Cell Metab 2022; 34:1654-1674. [PMID: 36323234 PMCID: PMC9641554 DOI: 10.1016/j.cmet.2022.10.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/23/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022]
Abstract
The evolution of glucagon has seen the transition from an impurity in the preparation of insulin to the development of glucagon receptor agonists for use in type 1 diabetes. In type 2 diabetes, glucagon receptor antagonists have been explored to reduce glycemia thought to be induced by hyperglucagonemia. However, the catabolic actions of glucagon are currently being leveraged to target the rise in obesity that paralleled that of diabetes, bringing the pharmacology of glucagon full circle. During this evolution, the physiological importance of glucagon advanced beyond the control of hepatic glucose production, incorporating critical roles for glucagon to regulate both lipid and amino acid metabolism. Thus, it is unsurprising that the study of glucagon has left several paradoxes that make it difficult to distill this hormone down to a simplified action. Here, we describe the history of glucagon from the past to the present and suggest some direction to the future of this field.
Collapse
Affiliation(s)
- Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA
| | - David A D'Alessio
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Endocrinology Division, Duke University Medical Center, Durham, NC 27701, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Endocrinology Division, Duke University Medical Center, Durham, NC 27701, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27701, USA.
| |
Collapse
|
20
|
Gola M, Sejda A, Godlewski J, Cieślak M, Starzyńska A. Neural Component of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:5246. [PMID: 36358664 PMCID: PMC9657005 DOI: 10.3390/cancers14215246] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/25/2022] [Indexed: 10/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive primary malignancy of the pancreas, with a dismal prognosis and limited treatment options. It possesses a unique tumor microenvironment (TME), generating dense stroma with complex elements cross-talking with each other to promote tumor growth and progression. Diversified neural components makes for not having a full understanding of their influence on its aggressive behavior. The aim of the study was to summarize and integrate the role of nerves in the pancreatic tumor microenvironment. The role of autonomic nerve fibers on PDAC development has been recently studied, which resulted in considering the targeting of sympathetic and parasympathetic pathways as a novel treatment opportunity. Perineural invasion (PNI) is commonly found in PDAC. As the severity of the PNI correlates with a poorer prognosis, new quantification of this phenomenon, distinguishing between perineural and endoneural invasion, could feature in routine pathological examination. The concepts of cancer-related neurogenesis and axonogenesis in PDAC are understudied; so, further research in this field may be warranted. A better understanding of the interdependence between the neural component and cancer cells in the PDAC microenvironment could bring new nerve-oriented treatment options into clinical practice and improve outcomes in patients with pancreatic cancer. In this review, we aim to summarize and integrate the current state of knowledge and future challenges concerning nerve-cancer interactions in PDAC.
Collapse
Affiliation(s)
- Michał Gola
- Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Aleksandra Sejda
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 18 Żołnierska Street, 10-561 Olsztyn, Poland
| | - Janusz Godlewski
- Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Małgorzata Cieślak
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 18 Żołnierska Street, 10-561 Olsztyn, Poland
| | - Anna Starzyńska
- Department of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland
| |
Collapse
|
21
|
Du YQ, Sha XY, Cheng J, Wang J, Lin JY, An WT, Pan W, Zhang LJ, Tao XN, Xu YF, Jia YL, Yang Z, Xiao P, Liu M, Sun JP, Yu X. Endogenous Lipid-GPR120 Signaling Modulates Pancreatic Islet Homeostasis to Different Extents. Diabetes 2022; 71:1454-1471. [PMID: 35472681 DOI: 10.2337/db21-0794] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022]
Abstract
Long-chain fatty acids (LCFAs) are not only energy sources but also serve as signaling molecules. GPR120, an LCFA receptor, plays key roles in maintaining metabolic homeostasis. However, whether endogenous ligand-GPR120 circuits exist and how such circuits function in pancreatic islets are unclear. Here, we found that endogenous GPR120 activity in pancreatic δ-cells modulated islet functions. At least two unsaturated LCFAs, oleic acid (OA) and linoleic acid (LA), were identified as GPR120 agonists within pancreatic islets. These two LCFAs promoted insulin secretion by inhibiting somatostatin secretion and showed bias activation of GPR120 in a model system. Compared with OA, LA exerted higher potency in promoting insulin secretion, which is dependent on β-arrestin2 function. Moreover, GPR120 signaling was impaired in the diabetic db/db model, and replenishing OA and LA improved islet function in both the db/db and streptozotocin-treated diabetic models. Consistently, the administration of LA improved glucose metabolism in db/db mice. Collectively, our results reveal that endogenous LCFA-GPR120 circuits exist and modulate homeostasis in pancreatic islets. The contributions of phenotype differences caused by different LCFA-GPR120 circuits within islets highlight the roles of fine-tuned ligand-receptor signaling networks in maintaining islet homeostasis.
Collapse
Affiliation(s)
- Ya-Qin Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xue-Ying Sha
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jie Cheng
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jing-Yu Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wen-Tao An
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wei Pan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Li-Jun Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao-Na Tao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Yun-Fei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ying-Li Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Peng Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
22
|
He Y, Fu Q, Sun M, Qian Y, Liang Y, Zhang J, Gao R, Jiang H, Dai H, Liu Y, Xu X, Chen H, Xu K, Yang T. Phosphoproteome reveals molecular mechanisms of aberrant rhythm in neurotransmitter-mediated islet hormone secretion in diabetic mice. Clin Transl Med 2022; 12:e890. [PMID: 35758323 PMCID: PMC9235066 DOI: 10.1002/ctm2.890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Acetylcholine (ACh) and norepinephrine (NE) are representative neurotransmitters of parasympathetic and sympathetic nerves, respectively, that antagonize each other to coregulate internal body functions. This also includes the control of different kinds of hormone secretion from pancreatic islets. However, the molecular mechanisms have not been fully elucidated, and whether innervation in islets is abnormal in diabetes mellitus also remains unclear. METHODS AND RESULTS Immunofluorescence colocalization and islet perfusion were performed and the results demonstrated that ACh/NE and their receptors were highly expressed in islet and rapidly regulated different hormones secretion. Phosphorylation is considered an important posttranslational modification in islet innervation and it was identified by quantitative proteomic and phosphoproteomic analyses in this study. The phosphorylated islet proteins were found involved in many biological and pathological processes, such as synaptic signalling transduction, calcium channel opening and insulin signalling pathway. Then, the kinases were predicted by motif analysis and further screened and verified by kinase-specific siRNAs in different islet cell lines (αTC1-6, Min6 and TGP52). After functional verification, Ksr2 and Pkacb were considered the key kinases of ACh and NE in insulin secretion, and Cadps, Mlxipl and Pdcd4 were the substrates of these kinases measured by immunofluorescence co-staining. Then, the decreased expression of receptors, kinases and substrates of ACh and NE were found in diabetic mice and the aberrant rhythm in insulin secretion could be improved by combined interventions on key receptors (M3 (pilocarpine) or α2a (guanfacine)) and kinases (Ksr2 or Pkacb). CONCLUSIONS Abnormal innervation was closely associated with the degree of islet dysfunction in diabetic mice and the aberrant rhythm in insulin secretion could be ameliorated significantly after intervention with key receptors and kinases in the early stage of diabetes mellitus, which may provide a promising therapeutic strategy for diabetes mellitus in the future.
Collapse
Affiliation(s)
- Yunqiang He
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Qi Fu
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Min Sun
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yu Qian
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yucheng Liang
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jie Zhang
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Rui Gao
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
- Oxford Centre for DiabetesEndocrinology and Metabolism, Radcliffe Department of Medicine, University of OxfordOxfordUK
| | - Hemin Jiang
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Hao Dai
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Yuwei Liu
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Xinyu Xu
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Heng Chen
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Kuanfeng Xu
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Tao Yang
- Department of Endocrinology and MetabolismThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| |
Collapse
|
23
|
Abdulrazzaq YM, Bastaki SMA, Adeghate E. Histamine H3 receptor antagonists - Roles in neurological and endocrine diseases and diabetes mellitus. Biomed Pharmacother 2022; 150:112947. [PMID: 35447544 DOI: 10.1016/j.biopha.2022.112947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/28/2022] [Accepted: 04/08/2022] [Indexed: 11/02/2022] Open
Abstract
Human histamine H3 receptor (H3R) was initially described in the brain of rat in 1983 and cloned in 1999. It can be found in the human brain and functions as a regulator of histamine synthesis and release. H3 receptors are predominantly resident in the presynaptic region of neurons containing histamine, where they modulate the synthesis and release of histamine (autoreceptor) or other neurotransmitters such as dopamine, norepinephrine, gamma-aminobutyric acid (GABA), glutamate, acetylcholine and serotonin (all heteroreceptors). The human histamine H3 receptor has twenty isoforms of which eight are functional. H3 receptor expression is seen in the cerebral cortex, neurons of the basal ganglia and hippocampus, which are important for process of cognition, sleep and homoeostatic regulation. In addition, histamine H3R antagonists stimulate insulin release, through inducing the release of acetylcholine and cause significant reduction in total body weight and triglycerides in obese subjects by causing a feeling of satiety in the hypothalamus. The ability of histamine H3R antagonist to reduce diabetes-induced hyperglycaemia is comparable to that of metformin. It is reasonable therefore, to claim that H3 receptor antagonists may play an important role in the therapy of disorders of cognition, the ability to sleep, oxidative stress, inflammation and anomaly of glucose homoeostasis. A large number of H3R antagonists are being developed by pharmaceutical companies and university research centres. As examples of these new drugs, this review will discuss a number of drugs, including the first histamine H3R receptor antagonist produced.
Collapse
Affiliation(s)
- Yousef M Abdulrazzaq
- Department of Paediatrics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Salim M A Bastaki
- Department of Pharmacology, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates; Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates.
| |
Collapse
|
24
|
Pan X, Tao S, Tong N. Potential Therapeutic Targeting Neurotransmitter Receptors in Diabetes. Front Endocrinol (Lausanne) 2022; 13:884549. [PMID: 35669692 PMCID: PMC9163348 DOI: 10.3389/fendo.2022.884549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Neurotransmitters are signaling molecules secreted by neurons to coordinate communication and proper function among different sections in the central neural system (CNS) by binding with different receptors. Some neurotransmitters as well as their receptors are found in pancreatic islets and are involved in the regulation of glucose homeostasis. Neurotransmitters can act with their receptors in pancreatic islets to stimulate or inhibit the secretion of insulin (β cell), glucagon (α cell) or somatostatin (δ cell). Neurotransmitter receptors are either G-protein coupled receptors or ligand-gated channels, their effects on blood glucose are mainly decided by the number and location of them in islets. Dysfunction of neurotransmitters receptors in islets is involved in the development of β cell dysfunction and type 2 diabetes (T2D).Therapies targeting different transmitter systems have great potential in the prevention and treatment of T2D and other metabolic diseases.
Collapse
Affiliation(s)
- Xiaohui Pan
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetes and Islet Transplantation, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Shibing Tao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology, Ziyang First People’s Hospital, Ziyang, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetes and Islet Transplantation, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Thor D. G protein-coupled receptors as regulators of pancreatic islet functionality. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119235. [PMID: 35151663 DOI: 10.1016/j.bbamcr.2022.119235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/03/2023]
Abstract
Glucose homeostasis is maintained by hormones secreted from different types of pancreatic islets and its dysregulation can result in diseases including diabetes mellitus. The secretion of hormones from pancreatic islets is highly complex and tightly controlled by G protein-coupled receptors (GPCRs). Moreover, GPCR signaling may play a role in enhancing islet cell replication and proliferation. Thus, targeting GPCRs offers a promising strategy for regulating the functionality of pancreatic islets. Here, available RNAseq datasets from human and mouse islets were used to identify the GPCR expression profile and the impact of GPCR signaling for normal islet functionality is discussed.
Collapse
Affiliation(s)
- Doreen Thor
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Johannisallee 30, 04103 Leipzig, Germany.
| |
Collapse
|
26
|
Hoyeck MP, Matteo G, MacFarlane EM, Perera I, Bruin JE. Persistent organic pollutants and β-cell toxicity: a comprehensive review. Am J Physiol Endocrinol Metab 2022; 322:E383-E413. [PMID: 35156417 PMCID: PMC9394781 DOI: 10.1152/ajpendo.00358.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 01/09/2023]
Abstract
Persistent organic pollutants (POPs) are a diverse family of contaminants that show widespread global dispersion and bioaccumulation. Humans are continuously exposed to POPs through diet, air particles, and household and commercial products; POPs are consistently detected in human tissues, including the pancreas. Epidemiological studies show a modest but consistent correlation between exposure to POPs and increased diabetes risk. The goal of this review is to provide an overview of epidemiological evidence and an in-depth evaluation of the in vivo and in vitro evidence that POPs cause β-cell toxicity. We review evidence for six classes of POPs: dioxins, polychlorinated biphenyls (PCBs), organochlorine pesticides (OCPs), organophosphate pesticides (OPPs), flame retardants, and per- and polyfluoroalkyl substances (PFAS). The available data provide convincing evidence implicating POPs as a contributing factor driving impaired glucose homeostasis, β-cell dysfunction, and altered metabolic and oxidative stress pathways in islets. These findings support epidemiological data showing that POPs increase diabetes risk and emphasize the need to consider the endocrine pancreas in toxicity assessments. Our review also highlights significant gaps in the literature assessing islet-specific endpoints after both in vivo and in vitro POP exposure. In addition, most rodent studies do not consider the impact of biological sex or secondary metabolic stressors in mediating the effects of POPs on glucose homeostasis and β-cell function. We discuss key gaps and limitations that should be assessed in future studies.
Collapse
Affiliation(s)
- Myriam P Hoyeck
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Geronimo Matteo
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Ontario, Canada
| | - Erin M MacFarlane
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Ineli Perera
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Jennifer E Bruin
- Department of Biology and Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
27
|
Guérineau NC, Campos P, Le Tissier PR, Hodson DJ, Mollard P. Cell Networks in Endocrine/Neuroendocrine Gland Function. Compr Physiol 2022; 12:3371-3415. [PMID: 35578964 DOI: 10.1002/cphy.c210031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reproduction, growth, stress, and metabolism are determined by endocrine/neuroendocrine systems that regulate circulating hormone concentrations. All these systems generate rhythms and changes in hormone pulsatility observed in a variety of pathophysiological states. Thus, the output of endocrine/neuroendocrine systems must be regulated within a narrow window of effective hormone concentrations but must also maintain a capacity for plasticity to respond to changing physiological demands. Remarkably most endocrinologists still have a "textbook" view of endocrine gland organization which has emanated from 20th century histological studies on thin 2D tissue sections. However, 21st -century technological advances, including in-depth 3D imaging of specific cell types have vastly changed our knowledge. We now know that various levels of multicellular organization can be found across different glands, that organizational motifs can vary between species and can be modified to enhance or decrease hormonal release. This article focuses on how the organization of cells regulates hormone output using three endocrine/neuroendocrine glands that present different levels of organization and complexity: the adrenal medulla, with a single neuroendocrine cell type; the anterior pituitary, with multiple intermingled cell types; and the pancreas with multiple intermingled cell types organized into distinct functional units. We give an overview of recent methodologies that allow the study of the different components within endocrine systems, particularly their temporal and spatial relationships. We believe the emerging findings about network organization, and its impact on hormone secretion, are crucial to understanding how homeostatic regulation of endocrine axes is carried out within endocrine organs themselves. © 2022 American Physiological Society. Compr Physiol 12:3371-3415, 2022.
Collapse
Affiliation(s)
| | - Pauline Campos
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Paul R Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,COMPARE University of Birmingham and University of Nottingham Midlands, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patrice Mollard
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
28
|
Moullé VS. Autonomic control of pancreatic beta cells: What is known on the regulation of insulin secretion and beta-cell proliferation in rodents and humans. Peptides 2022; 148:170709. [PMID: 34896576 DOI: 10.1016/j.peptides.2021.170709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 11/21/2022]
Abstract
Insulin secretion and pancreatic beta-cell proliferation are tightly regulated by several signals such as hormones, nutrients, and neurotransmitters. However, the autonomic control of beta cells is not fully understood. In this review, we describe mechanisms involved in insulin secretion as well as metabolic and mitogenic actions on its target tissues. Since pancreatic islets are physically connected to the brain by nerves, parasympathetic and sympathetic neurotransmitters can directly potentiate or repress insulin secretion and beta-cell proliferation. Finally, we highlight the role of the autonomic nervous system in metabolic diseases such as diabetes and obesity.
Collapse
|
29
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:973-984. [DOI: 10.1093/jpp/rgac021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022]
|
30
|
Langlois A, Dumond A, Vion J, Pinget M, Bouzakri K. Crosstalk Communications Between Islets Cells and Insulin Target Tissue: The Hidden Face of Iceberg. Front Endocrinol (Lausanne) 2022; 13:836344. [PMID: 35185804 PMCID: PMC8851682 DOI: 10.3389/fendo.2022.836344] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
The regulation of insulin secretion is under control of a complex inter-organ/cells crosstalk involving various metabolites and/or physical connections. In this review, we try to illustrate with current knowledge how β-cells communicate with other cell types and organs in physiological and pathological contexts. Moreover, this review will provide a better understanding of the microenvironment and of the context in which β-cells exist and how this can influence their survival and function. Recent studies showed that β-cell insulin secretion is regulated also by a direct and indirect inter-organ/inter-cellular communication involving various factors, illustrating the idea of "the hidden face of the iceberg". Moreover, any disruption on the physiological communication between β-cells and other cells or organs can participate on diabetes onset. Therefore, for new anti-diabetic treatments' development, it is necessary to consider the entire network of cells and organs involved in the regulation of β-cellular function and no longer just β-cell or pancreatic islet alone. In this context, we discuss here the intra-islet communication, the β-cell/skeletal muscle, β-cell/adipose tissue and β-cell/liver cross talk.
Collapse
|
31
|
Chung JY, Kim OY, Song J. Role of ketone bodies in diabetes-induced dementia: sirtuins, insulin resistance, synaptic plasticity, mitochondrial dysfunction, and neurotransmitter. Nutr Rev 2021; 80:774-785. [PMID: 34957519 PMCID: PMC8907488 DOI: 10.1093/nutrit/nuab118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abstract
Patients with type 2 diabetes can have several neuropathologies, such as memory deficits. Recent studies have focused on the association between metabolic imbalance and neuropathological problems, and the associated molecular pathology. Diabetes triggers neuroinflammation, impaired synaptic plasticity, mitochondrial dysfunction, and insulin resistance in the brain. Glucose is a main energy substrate for neurons, but under certain conditions, such as fasting and starvation, ketone bodies can be used as an energy fuel for these cells. Recent evidence has shed new light on the role of ketone bodies in regulating several anti-inflammation cellular pathways and improving glucose metabolism, insulin action, and synaptic plasticity, thereby being neuroprotective. However, very high amount of ketone bodies can be toxic for the brain, such as in ketoacidosis, a dangerous complication that may occur in type 1 diabetes mellitus or alcoholism. Recent findings regarding the relationship between ketone bodies and neuropathogenesis in dementia are reviewed in this article. They suggest that the adequately low amount of ketone bodies can be a potential energy source for the treatment of diabetes-induced dementia neuropathology, considering the multifaceted effects of the ketone bodies in the central nervous system. This review can provide useful information for establishing the therapeutic guidelines of a ketogenic diet for diabetes-induced dementia.
Collapse
Affiliation(s)
- Ji Yeon Chung
- Department of Neurology, Chosun University Medical School, Gwangju, Republic of Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition and the Department of Health Sciences, Dong-A University, Busan, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Jeollanam-do, Republic of Korea
| |
Collapse
|
32
|
Panzer JK, Caicedo A. Targeting the Pancreatic α-Cell to Prevent Hypoglycemia in Type 1 Diabetes. Diabetes 2021; 70:2721-2732. [PMID: 34872936 PMCID: PMC8660986 DOI: 10.2337/dbi20-0048] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022]
Abstract
Life-threatening hypoglycemia is a limiting factor in the management of type 1 diabetes. People with diabetes are prone to develop hypoglycemia because they lose physiological mechanisms that prevent plasma glucose levels from falling. Among these so-called counterregulatory responses, secretion of glucagon from pancreatic α-cells is preeminent. Glucagon, a hormone secreted in response to a lowering in glucose concentration, counteracts a further drop in glycemia by promoting gluconeogenesis and glycogenolysis in target tissues. In diabetes, however, α-cells do not respond appropriately to changes in glycemia and, thus, cannot mount a counterregulatory response. If the α-cell could be targeted therapeutically to restore its ability to prevent hypoglycemia, type 1 diabetes could be managed more efficiently and safely. Unfortunately, the mechanisms that allow the α-cell to respond to hypoglycemia have not been fully elucidated. We know even less about the pathophysiological mechanisms that cause α-cell dysfunction in diabetes. Based on published findings and unpublished observations, and taking into account its electrophysiological properties, we propose here a model of α-cell function that could explain its impairment in diabetes. Within this frame, we emphasize those elements that could be targeted pharmacologically with repurposed U.S. Food and Drug Administration-approved drugs to rescue α-cell function and restore glucose counterregulation in people with diabetes.
Collapse
Affiliation(s)
- Julia K Panzer
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
33
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
34
|
Henquin JC. Non-glucose modulators of insulin secretion in healthy humans: (dis)similarities between islet and in vivo studies. Metabolism 2021; 122:154821. [PMID: 34174327 DOI: 10.1016/j.metabol.2021.154821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022]
Abstract
Optimal metabolic homeostasis requires precise temporal and quantitative control of insulin secretion. Both in vivo and in vitro studies have often focused on the regulation by glucose although many additional factors including other nutrients, neurotransmitters, hormones and drugs, modulate the secretory function of pancreatic β-cells. This review is based on the analysis of clinical investigations characterizing the effects of non-glucose modulators of insulin secretion in healthy subjects, and of experimental studies testing the same modulators in islets isolated from normal human donors. The aim was to determine whether the information gathered in vitro can reliably be translated to the in vivo situation. The comparison evidenced both convincing similarities and areas of discordance. The lack of coherence generally stems from the use of exceedingly high concentrations of test agents at too high or too low glucose concentrations in vitro, which casts doubts on the physiological relevance of a number of observations made in isolated islets. Future projects resorting to human islets should avoid extreme experimental conditions, such as oversized stimulations or inhibitions of β-cells, which are unlikely to throw light on normal insulin secretion and contribute to the elucidation of its defects.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
35
|
Lugo-Fabres PH, Otero-Sastre LM, Bernáldez-Sarabia J, Camacho-Villegas TA, Sánchez-Campos N, Serrano-Bello J, Medina LA, Muñiz-Hernández S, de la Cruz L, Arenas I, Barajas-Martínez A, Garcia DE, Nuñez-Garcia L, González-Canudas J, Licea-Navarro AF. Potential Therapeutic Applications of Synthetic Conotoxin s-cal14.2b, Derived from Californiconus californicus, for Treating Type 2 Diabetes. Biomedicines 2021; 9:936. [PMID: 34440140 PMCID: PMC8391312 DOI: 10.3390/biomedicines9080936] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/25/2021] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
The FDA's approval of peptide drugs such as Ziconotide or Exendin for pain relief and diabetes treatment, respectively, enhanced the interest to explore novel conotoxins from Conus species venom. In general, conotoxins can be used in pathologies where voltage-gated channels, membrane receptors, or ligands alter normal physiological functions, as in metabolic diseases such as Type 2 diabetes. In this study, the synthetic cal14.2b (s-cal14.2b) from the unusual Californiconus californicus demonstrated bioactivity on NIT-1 insulinoma cell lines stimulating insulin secretion detecting by high performance liquid chromatography (HPLC). Accordingly, s-cal14.2b increased the CaV1.2/1.3 channel-current by 35 ± 4% with a recovery τ of 10.3 ± 4 s in primary cell culture of rat pancreatic β-cells. The in vivo results indicated a similar effect of insulin secretion on mice in the glucose tolerance curve model by reducing the glucose from 500 mg/dL to 106 mg/dL in 60 min, compared to the negative control of 325 mg/dL at the same time. The PET-SCAN with radiolabeling 99mTc-s-cal14.2b demonstrated biodistribution and accumulation in rat pancreas with complete depuration in 24 h. These findings show the potential therapeutic use of s-cal14.2b in endocrinal pathologies such as early stages of Type 2 Diabetes where the pancreas's capability to produce insulin is still effective.
Collapse
Affiliation(s)
- Pavel H. Lugo-Fabres
- CONACYT-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ) A. C., Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico; (P.H.L.-F.); (T.A.C.-V.)
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, Ensenada 22860, Baja California, Mexico; (L.M.O.-S.); (J.B.-S.); (N.S.-C.)
| | - Leslie M. Otero-Sastre
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, Ensenada 22860, Baja California, Mexico; (L.M.O.-S.); (J.B.-S.); (N.S.-C.)
| | - Johanna Bernáldez-Sarabia
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, Ensenada 22860, Baja California, Mexico; (L.M.O.-S.); (J.B.-S.); (N.S.-C.)
| | - Tanya A. Camacho-Villegas
- CONACYT-Unidad de Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ) A. C., Av. Normalistas 800, Colinas de la Normal, Guadalajara 44270, Jalisco, Mexico; (P.H.L.-F.); (T.A.C.-V.)
| | - Noemi Sánchez-Campos
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, Ensenada 22860, Baja California, Mexico; (L.M.O.-S.); (J.B.-S.); (N.S.-C.)
| | - Janeth Serrano-Bello
- Laboratorio de Bioingeniería de Tejidos, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Ciudad de México 04360, Mexico;
| | - Luis A. Medina
- Laboratorio de Física Médica-Unidad de Investigación Biomédica en Cáncer-INCan, Ciudad de México 14080, Mexico;
- Instituto de Física, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico
| | - Saé Muñiz-Hernández
- Laboratorio de Oncología Experimental, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Ciudad de México 14080, Mexico;
| | - Lizbeth de la Cruz
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.d.l.C.); (I.A.); (A.B.-M.); (D.E.G.)
| | - Isabel Arenas
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.d.l.C.); (I.A.); (A.B.-M.); (D.E.G.)
| | - Antonio Barajas-Martínez
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.d.l.C.); (I.A.); (A.B.-M.); (D.E.G.)
| | - David E. Garcia
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico; (L.d.l.C.); (I.A.); (A.B.-M.); (D.E.G.)
| | - Linda Nuñez-Garcia
- Laboratorios Silanes S.A. de C.V., Ciudad de México 11000, Mexico; (L.N.-G.); (J.G.-C.)
| | | | - Alexei F. Licea-Navarro
- Departamento de Innovación Biomédica, Centro de Investigación Científica y de Educación Superior de Ensenada (CICESE), Carretera Ensenada-Tijuana No. 3918, Zona Playitas, Ensenada 22860, Baja California, Mexico; (L.M.O.-S.); (J.B.-S.); (N.S.-C.)
| |
Collapse
|
36
|
Lkhagvasuren B, Mee-Inta O, Zhao ZW, Hiramoto T, Boldbaatar D, Kuo YM. Pancreas-Brain Crosstalk. Front Neuroanat 2021; 15:691777. [PMID: 34354571 PMCID: PMC8329585 DOI: 10.3389/fnana.2021.691777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
The neural regulation of glucose homeostasis in normal and challenged conditions involves the modulation of pancreatic islet-cell function. Compromising the pancreas innervation causes islet autoimmunity in type 1 diabetes and islet cell dysfunction in type 2 diabetes. However, despite the richly innervated nature of the pancreas, islet innervation remains ill-defined. Here, we review the neuroanatomical and humoral basis of the cross-talk between the endocrine pancreas and autonomic and sensory neurons. Identifying the neurocircuitry and neurochemistry of the neuro-insular network would provide clues to neuromodulation-based approaches for the prevention and treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Battuvshin Lkhagvasuren
- Brain Science Institute, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Onanong Mee-Inta
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Zi-Wei Zhao
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Tetsuya Hiramoto
- Department of Psychosomatic Medicine, Fukuoka Hospital, National Hospital Organization, Fukuoka, Japan
| | - Damdindorj Boldbaatar
- Brain Science Institute, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan.,Department of Cell Biology and Anatomy, National Cheng Kung University College of Medicine, Tainan, Taiwan
| |
Collapse
|
37
|
Ng XW, Chung YH, Piston DW. Intercellular Communication in the Islet of Langerhans in Health and Disease. Compr Physiol 2021; 11:2191-2225. [PMID: 34190340 PMCID: PMC8985231 DOI: 10.1002/cphy.c200026] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Blood glucose homeostasis requires proper function of pancreatic islets, which secrete insulin, glucagon, and somatostatin from the β-, α-, and δ-cells, respectively. Each islet cell type is equipped with intrinsic mechanisms for glucose sensing and secretory actions, but these intrinsic mechanisms alone cannot explain the observed secretory profiles from intact islets. Regulation of secretion involves interconnected mechanisms among and between islet cell types. Islet cells lose their normal functional signatures and secretory behaviors upon dispersal as compared to intact islets and in vivo. In dispersed islet cells, the glucose response of insulin secretion is attenuated from that seen from whole islets, coordinated oscillations in membrane potential and intracellular Ca2+ activity, as well as the two-phase insulin secretion profile, are missing, and glucagon secretion displays higher basal secretion profile and a reverse glucose-dependent response from that of intact islets. These observations highlight the critical roles of intercellular communication within the pancreatic islet, and how these communication pathways are crucial for proper hormonal and nonhormonal secretion and glucose homeostasis. Further, misregulated secretions of islet secretory products that arise from defective intercellular islet communication are implicated in diabetes. Intercellular communication within the islet environment comprises multiple mechanisms, including electrical synapses from gap junctional coupling, paracrine interactions among neighboring cells, and direct cell-to-cell contacts in the form of juxtacrine signaling. In this article, we describe the various mechanisms that contribute to proper islet function for each islet cell type and how intercellular islet communications are coordinated among the same and different islet cell types. © 2021 American Physiological Society. Compr Physiol 11:2191-2225, 2021.
Collapse
Affiliation(s)
- Xue W Ng
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - Yong H Chung
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| |
Collapse
|
38
|
Sluga N, Postić S, Sarikas S, Huang YC, Stožer A, Slak Rupnik M. Dual Mode of Action of Acetylcholine on Cytosolic Calcium Oscillations in Pancreatic Beta and Acinar Cells In Situ. Cells 2021; 10:1580. [PMID: 34201461 PMCID: PMC8305080 DOI: 10.3390/cells10071580] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cholinergic innervation in the pancreas controls both the release of digestive enzymes to support the intestinal digestion and absorption, as well as insulin release to promote nutrient use in the cells of the body. The effects of muscarinic receptor stimulation are described in detail for endocrine beta cells and exocrine acinar cells separately. Here we describe morphological and functional criteria to separate these two cell types in situ in tissue slices and simultaneously measure their response to ACh stimulation on cytosolic Ca2+ oscillations [Ca2+]c in stimulatory glucose conditions. Our results show that both cell types respond to glucose directly in the concentration range compatible with the glucose transporters they express. The physiological ACh concentration increases the frequency of glucose stimulated [Ca2+]c oscillations in both cell types and synchronizes [Ca2+]c oscillations in acinar cells. The supraphysiological ACh concentration further increases the oscillation frequency on the level of individual beta cells, inhibits the synchronization between these cells, and abolishes oscillatory activity in acinar cells. We discuss possible mechanisms leading to the observed phenomena.
Collapse
Affiliation(s)
- Nastja Sluga
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (N.S.); (A.S.)
| | - Sandra Postić
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
| | - Srdjan Sarikas
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
| | - Ya-Chi Huang
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (N.S.); (A.S.)
| | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (N.S.); (A.S.)
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
- Alma Mater Europaea, European Center Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
39
|
Abdulreda MH, Berggren PO. The pancreatic islet: a micro-organ in control. CELLR4-- REPAIR, REPLACEMENT, REGENERATION, & REPROGRAMMING 2021; 9. [PMID: 33816651 PMCID: PMC8017525 DOI: 10.32113/cellr4_20213_3093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The islets of Langerhans constitute the endocrine pancreas which regulates blood glucose homeostasis and their dysfunction results in diabetes. Each of the pancreatic islets constitutes an entire micro-organ with intricate cell to cell interactions and that is well vascularized and innervated. An important therapeutic advantage in islet transplant is that pancreatic islets maintain their organ integrity when isolated and transplanted to patients with severe diabetes. Once transplanted, the islet micro-organs actively contribute to their own vascularization and start to function immediately. Hence, in terms of organ transplantation, the application of pancreatic islets will be a decisive clinical tool for future diabetes care (credit: Tilo Moede).
Collapse
Affiliation(s)
- M H Abdulreda
- Department of Surgery, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - P O Berggren
- Department of Surgery, Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, SE-17176 Stockholm, Sweden
| |
Collapse
|
40
|
Fontaine AK, Ramirez DG, Littich SF, Piscopio RA, Kravets V, Schleicher WE, Mizoguchi N, Caldwell JH, Weir RFF, Benninger RKP. Optogenetic stimulation of cholinergic fibers for the modulation of insulin and glycemia. Sci Rep 2021; 11:3670. [PMID: 33574598 PMCID: PMC7878862 DOI: 10.1038/s41598-021-83361-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/01/2021] [Indexed: 01/12/2023] Open
Abstract
Previous studies have demonstrated stimulation of endocrine pancreas function by vagal nerve electrical stimulation. While this increases insulin secretion, expected concomitant reductions in circulating glucose do not occur. A complicating factor is the non-specific nature of electrical nerve stimulation. Optogenetic tools, however, provide the potential for cell-type specific neural stimulation using genetic targeting and/or spatially shaped excitation light. Here, we demonstrate light-activated stimulation of the endocrine pancreas by targeting parasympathetic (cholinergic) axons. In a mouse model expressing ChannelRhodopsin2 (ChR2) in cholinergic cells, serum insulin and glucose were measured in response to (1) ultrasound image-guided optical stimulation of axon terminals in the pancreas or (2) optical stimulation of axons of the cervical vagus nerve. Measurements were made in basal-glucose and glucose-stimulated conditions. Significant increases in plasma insulin occurred relative to controls under both pancreas and cervical vagal stimulation, while a rapid reduction in glycemic levels were observed under pancreatic stimulation. Additionally, ultrasound-based measurements of blood flow in the pancreas were increased under pancreatic stimulation. Together, these results demonstrate the utility of in-vivo optogenetics for studying the neural regulation of endocrine pancreas function and suggest its therapeutic potential for the control of insulin secretion and glucose homeostasis.
Collapse
Affiliation(s)
- Arjun K Fontaine
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Boulder, USA.
- Biomechatronics Development Laboratory, University of Colorado, Anschutz Medical Campus, Boulder, USA.
| | - David G Ramirez
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Boulder, USA
- Barbara Davis Center for Childhood Diabetes - Anschutz Medical Campus, Boulder, USA
| | - Samuel F Littich
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Boulder, USA
- Biomechatronics Development Laboratory, University of Colorado, Anschutz Medical Campus, Boulder, USA
| | - Robert A Piscopio
- Barbara Davis Center for Childhood Diabetes - Anschutz Medical Campus, Boulder, USA
| | - Vira Kravets
- Barbara Davis Center for Childhood Diabetes - Anschutz Medical Campus, Boulder, USA
| | | | - Naoko Mizoguchi
- Division of Pharmacology, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, Saitama, Japan
| | - John H Caldwell
- Department of Cell and Developmental Biology, University of Colorado, Anschutz Medical Campus, Boulder, USA
| | - Richard F Ff Weir
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Boulder, USA
- Biomechatronics Development Laboratory, University of Colorado, Anschutz Medical Campus, Boulder, USA
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado, Anschutz Medical Campus, Boulder, USA.
- Barbara Davis Center for Childhood Diabetes - Anschutz Medical Campus, Boulder, USA.
| |
Collapse
|
41
|
Gao R, Yang T, Zhang Q. δ-Cells: The Neighborhood Watch in the Islet Community. BIOLOGY 2021; 10:biology10020074. [PMID: 33494193 PMCID: PMC7909827 DOI: 10.3390/biology10020074] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 12/29/2022]
Abstract
Somatostatin-secreting δ-cells have aroused great attention due to their powerful roles in coordination of islet insulin and glucagon secretion and maintenance of glucose homeostasis. δ-cells exhibit neuron-like morphology with projections which enable pan-islet somatostatin paracrine regulation despite their scarcity in the islets. The expression of a range of hormone and neurotransmitter receptors allows δ-cells to integrate paracrine, endocrine, neural and nutritional inputs, and provide rapid and precise feedback modulations on glucagon and insulin secretion from α- and β-cells, respectively. Interestingly, the paracrine tone of δ-cells can be effectively modified in response to factors released by neighboring cells in this interactive communication, such as insulin, urocortin 3 and γ-aminobutyric acid from β-cells, glucagon, glutamate and glucagon-like peptide-1 from α-cells. In the setting of diabetes, defects in δ-cell function lead to suboptimal insulin and glucagon outputs and lift the glycemic set-point. The interaction of δ-cells and non-δ-cells also becomes defective in diabetes, with reduces paracrine feedback to β-cells to exacerbate hyperglycemia or enhanced inhibition of α-cells, disabling counter-regulation, to cause hypoglycemia. Thus, it is possible to restore/optimize islet function in diabetes targeting somatostatin signaling, which could open novel avenues for the development of effective diabetic treatments.
Collapse
Affiliation(s)
- Rui Gao
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK;
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| | - Quan Zhang
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK;
- Correspondence:
| |
Collapse
|
42
|
Henquin JC. Paracrine and autocrine control of insulin secretion in human islets: evidence and pending questions. Am J Physiol Endocrinol Metab 2021; 320:E78-E86. [PMID: 33103455 DOI: 10.1152/ajpendo.00485.2020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Insulin secretion by β-cells is largely controlled by circulating nutrients, hormones, and neurotransmitters. However, recent years have witnessed the multiplication of studies investigating whether local regulation also takes place within pancreatic islets, in which β-cells cohabit with several other cell types. The cell composition and architectural organization of human islets differ from those of rodent islets and are particularly favorable to cellular interactions. An impressive number of hormonal (glucagon, glucagon-like peptide-1, somatostatin, etc.) and nonhormonal products (ATP, acetylcholine, γ-aminobutyric acid, dopamine, etc.) are released by islet cells and have been implicated in a local control of insulin secretion. This review analyzes reports directly testing paracrine and autocrine control of insulin secretion in isolated human islets. Many of these studies were designed on background information collected in rodent islets. However, the perspective of the review is not to highlight species similarities or specificities but to contrast established and speculative mechanisms in human islets. It will be shown that the current evidence is convincing only for a minority of candidates for a paracrine function whereas arguments supporting a physiological role of others do not stand up to scrutiny. Several pending questions await further investigation.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium
| |
Collapse
|
43
|
Weitz J, Menegaz D, Caicedo A. Deciphering the Complex Communication Networks That Orchestrate Pancreatic Islet Function. Diabetes 2021; 70:17-26. [PMID: 33355306 PMCID: PMC7881851 DOI: 10.2337/dbi19-0033] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/01/2020] [Indexed: 12/27/2022]
Abstract
Pancreatic islets are clusters of hormone-secreting endocrine cells that rely on intricate cell-cell communication mechanisms for proper function. The importance of multicellular cooperation in islet cell physiology was first noted nearly 30 years ago in seminal studies showing that hormone secretion from endocrine cell types is diminished when these cells are dispersed. These studies showed that reestablishing cellular contacts in so-called pseudoislets caused endocrine cells to regain hormone secretory function. This not only demonstrated that cooperation between islet cells is highly synergistic but also gave birth to the field of pancreatic islet organoids. Here we review recent advances related to the mechanisms of islet cell cross talk. We first describe new developments that revise current notions about purinergic and GABA signaling in islets. Then we comment on novel multicellular imaging studies that are revealing emergent properties of islet communication networks. We finish by highlighting and discussing recent synthetic approaches that use islet organoids of varied cellular composition to interrogate intraislet signaling mechanisms. This reverse engineering of islets not only will shed light on the mechanisms of intraislet signaling and define communication networks but also may guide efforts aimed at restoring islet function and β-cell mass in diabetes.
Collapse
Affiliation(s)
- Jonathan Weitz
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | - Danusa Menegaz
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Leonard M. Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Leonard M. Miller School of Medicine, Miami, FL
| |
Collapse
|
44
|
Sanabria V, Bittencourt S, Perosa SR, de la Rosa T, da Graça Naffah-Mazzacoratti M, Andersen ML, Tufik S, Cavalheiro EA, Amado D. Hormonal and biochemical changes in female Proechimys guyannensis, an animal model of resistance to pilocarpine-induced status epilepticus. Sci Rep 2020; 10:20982. [PMID: 33268798 PMCID: PMC7710747 DOI: 10.1038/s41598-020-77879-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 11/10/2020] [Indexed: 11/09/2022] Open
Abstract
The Amazon rodent Proechimys guyannensis is widely studied for hosting various pathogens, though rarely getting sick. Previous studies on male Proechimys have revealed an endogenous resistance to epilepsy. Here, we assess in female Proechimys, whether sex hormones and biochemical aspects can interfere with the induction of status epilepticus (SE). The lithium-pilocarpine ramp-up protocol was used to induce SE, and blood sera were collected at 30 and 90 min after SE, alongside brains, for biochemical, western blot and immunohistochemical analyses. Results from non-ovariectomised (NOVX) Proechimys were compared to ovariectomised (OVX) animals. Data from female Wistars were used as a positive control of SE inductions. SE latency was similar in NOVX, OVX, and female Wistars groups. However, the pilocarpine dose required to induce SE in Proechimys was higher (25- to 50-folds more). Despite a higher dose, Proechimys did not show strong SE like Wistars; they only reached stage 2 of the Racine scale. These data suggest that female Proechimys are resistant to SE induction. Glucose and progesterone levels increased at 30 min and returned to normal at 90 min after SE. A relevant fact because in humans and rodents, SE leads to hypoglycaemia after 30 min of SE and does not return to normal levels in a short time, a typical adverse effect of SE. In OVX animals, a decrease in GABAergic receptors within 90 min of SE may suggest that ovariectomy produces changes in the hippocampus, including a certain vulnerability to seizures. We speculate that progesterone and glucose increases form part of the compensatory mechanisms that provide resistance in Proechimys against SE induction.
Collapse
Affiliation(s)
- Viviam Sanabria
- Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, São Paulo, SP, Brazil.
| | - Simone Bittencourt
- Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, São Paulo, SP, Brazil
| | - Sandra R Perosa
- Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, São Paulo, SP, Brazil
| | - Tomás de la Rosa
- Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, São Paulo, SP, Brazil
| | | | - Monica L Andersen
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 826, São Paulo, SP, Brazil
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), Rua Botucatu, 826, São Paulo, SP, Brazil
| | - Esper A Cavalheiro
- Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, São Paulo, SP, Brazil
| | - Débora Amado
- Department of Neurology and Neurosurgery, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo, 669, São Paulo, SP, Brazil
| |
Collapse
|
45
|
The Molecular Mechanisms by Which Vitamin D Prevents Insulin Resistance and Associated Disorders. Int J Mol Sci 2020; 21:ijms21186644. [PMID: 32932777 PMCID: PMC7554927 DOI: 10.3390/ijms21186644] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Numerous studies have shown that vitamin D deficiency is very common in modern societies and is perceived as an important risk factor in the development of insulin resistance and related diseases such as obesity and type 2 diabetes (T2DM). While it is generally accepted that vitamin D is a regulator of bone homeostasis, its ability to counteract insulin resistance is subject to debate. The goal of this communication is to review the molecular mechanism by which vitamin D reduces insulin resistance and related complications. The university library, PUBMED, and Google Scholar were searched to find relevant studies to be summarized in this review article. Insulin resistance is accompanied by chronic hyperglycaemia and inflammation. Recent studies have shown that vitamin D exhibits indirect antioxidative properties and participates in the maintenance of normal resting ROS level. Appealingly, vitamin D reduces inflammation and regulates Ca2+ level in many cell types. Therefore, the beneficial actions of vitamin D include diminished insulin resistance which is observed as an improvement of glucose and lipid metabolism in insulin-sensitive tissues.
Collapse
|
46
|
Selig DJ, Brown AJ, DeLuca JP, Kress AT, Livezey JR, Oliver TG, Por ED, Thelus Jean R. Risk of type 2 diabetes mellitus by antimuscarinic agents among adult females receiving care in the military health system. Pharmacoepidemiol Drug Saf 2020; 29:1605-1615. [PMID: 32897626 DOI: 10.1002/pds.5090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/09/2020] [Accepted: 07/11/2020] [Indexed: 11/05/2022]
Abstract
PURPOSE To explore patterns of antimuscarinic medication as a risk factor for type 2 diabetes mellitus (T2DM). METHODS This is a retrospective cohort study of females 18 years or older within the Military Health System from 2006 to 2016. Administrative and claims data were used to select patients who initiated therapy with tolterodine, fesoterodine, oxybutynin, darifenacin, solifenacin, or trospium. Patients with no documented history of T2DM were followed for the occurrence of T2DM, the end of the study or loss of eligibility. Rates of T2DM were calculated for the overall population, by duration of therapy and by individual drugs. Crude and adjusted Cox proportional hazards were calculated to assess differences by duration of use and specific muscarinic antagonist. RESULTS Over 2.6 million antimuscarinic prescriptions were dispensed to 241 829 females (mean age/SD, 62 ± 18 years). Patients exposed to M3 selective antagonists had highest risk of developing T2DM compared to those exposed to nonselective antagonists. Using oxybutynin, a nonselective antagonist as a comparator, adjusted rate ratios of T2DM were 57% (HR 1.57, 95%CI 1.48-1.67) and 29% (HR 1.29, 95%CI 1.24-1.35) significantly higher for darifenacin and solifenacin, respectively (both M3 selective). CONCLUSIONS We found exposure to M3 selective antagonists darifenacin and solifenacin had the highest risk of developing T2DM compared to nonselective antagonist oxybutynin. This is supported by well described physiologic mechanisms and may allow for more informed prescribing decisions, particularly if minimizing risk of T2DM is a priority.
Collapse
Affiliation(s)
- Daniel J Selig
- Walter Reed Army Institute of Research, Experimental Therapeutics, 503 Robert Grant Ave, Silver Spring, Maryland, 20910, USA
| | - Aaron J Brown
- Uniformed Services University, Department of Medicine, Division of Clinical Pharmacology, 4301 Jones Bridge Rd, Bethesda, Maryland, 20814, USA
| | - Jesse P DeLuca
- Walter Reed Army Institute of Research, Experimental Therapeutics, 503 Robert Grant Ave, Silver Spring, Maryland, 20910, USA
| | - Adrian T Kress
- Army Office of the Surgeon General, Pharmacovigilance Center, 7700 Arlington Boulevard, Falls Church, Virginia, 22042, USA
| | - Jeffrey R Livezey
- Uniformed Services University, Department of Medicine, Division of Clinical Pharmacology, 4301 Jones Bridge Rd, Bethesda, Maryland, 20814, USA
| | - Thomas G Oliver
- Uniformed Services University, Department of Medicine, Division of Clinical Pharmacology, 4301 Jones Bridge Rd, Bethesda, Maryland, 20814, USA
| | - Elaine D Por
- Walter Reed Army Institute of Research, Experimental Therapeutics, 503 Robert Grant Ave, Silver Spring, Maryland, 20910, USA
| | - Rosenie Thelus Jean
- Army Office of the Surgeon General, Pharmacovigilance Center, 7700 Arlington Boulevard, Falls Church, Virginia, 22042, USA
| |
Collapse
|
47
|
Mussa BM, Srivastava A, Mohammed AK, Verberne AJM. Nitric oxide interacts with cholinoceptors to modulate insulin secretion by pancreatic β cells. Pflugers Arch 2020; 472:1469-1480. [PMID: 32803305 PMCID: PMC7476970 DOI: 10.1007/s00424-020-02443-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/17/2020] [Accepted: 07/31/2020] [Indexed: 01/09/2023]
Abstract
Dysfunction of the pancreatic β cells leads to several chronic disorders including diabetes mellitus. Several mediators and mechanisms are known to be involved in the regulation of β cell secretory function. In this study, we propose that cytokine-induced nitric oxide (NO) production interacts with cholinergic mechanisms to modulate insulin secretion from pancreatic β cells. Using a rat insulinoma cell line INS-1, we demonstrated that β cell viability decreases significantly in the presence of SNAP (NO donor) in a concentration- and time-dependent manner. Cell viability was also found to be decreased in the presence of a combined treatment of SNAP with SMN (muscarinic receptor antagonist). We then investigated the impact of these findings on insulin secretion and found a significant reduction in glucose uptake by INS-1 cells in the presence of SNAP and SMN as compared with control. Nitric oxide synthase 3 gene expression was found to be significantly reduced in response to combined treatment with SNAP and SMN suggesting an interaction between the cholinergic and nitrergic systems. The analysis of gene and protein expression further pin-pointed the involvement of M3 muscarinic receptors in the cholinergic pathway. Upon treatment with cytokines, reduced cell viability was observed in the presence of TNF-α and IFN-γ. A significant reduction in insulin secretion was also noted after treatment with TNF-α and IFN-γ and IL1-β. The findings of the present study have shown for the first time that the inhibition of the excitatory effects of cholinergic pathways on glucose-induced insulin secretion may cause β cell injury and dysfunction of insulin secretion in response to cytokine-induced NO production.
Collapse
Affiliation(s)
- Bashair M Mussa
- Basic Medical Science Department, College of Medicine, University of Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates.
| | - Ankita Srivastava
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Abdul Khader Mohammed
- Sharjah Institute for Medical Research, University of Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Anthony J M Verberne
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, 3084, Australia
| |
Collapse
|
48
|
Pralle RS, Schultz NE, White HM, Weigel KA. Hyperketonemia GWAS and parity-dependent SNP associations in Holstein dairy cows intensively sampled for blood β-hydroxybutyrate concentration. Physiol Genomics 2020; 52:347-357. [PMID: 32628084 DOI: 10.1152/physiolgenomics.00016.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hyperketonemia (HYK) is a metabolic disorder that affects early postpartum dairy cows; however, there has been limited success in identifying genomic variants contributing to HYK susceptibility. We conducted a genome-wide association study (GWAS) using HYK phenotypes based on an intensive screening protocol, interrogated genotype interactions with parity group (GWIS), and evaluated the enrichment of annotated metabolic pathways. Holstein cows were enrolled into the experiment after parturition, and blood samples were collected at four timepoints between 5 and 18 days postpartum. Concentration of blood β-hydroxybutyrate (BHB) was quantified cow-side via a handheld BHB meter. Cows were labeled as a HYK case when at least one blood sample had BHB ≥ 1.2 mmol/L, and all other cows were considered non-HYK controls. After quality control procedures, 1,710 cows and 58,699 genotypes were available for further analysis. The GWAS and GWIS were performed using the forward feature select linear mixed model method. There was evidence for an association between ARS-BFGL-NGS-91238 and HYK susceptibility, as well as parity-dependent associations to HYK for BovineHD0600024247 and BovineHD1400023753. Candidate genes annotated to these single nuclear polymorphism associations have been previously associated with obesity, diabetes, insulin resistance, and fatty liver in humans and rodent models. Enrichment analysis revealed focal adhesion and axon guidance as metabolic pathways contributing to HYK etiology, while genetic variation in pathways related to insulin secretion and sensitivity may affect HYK susceptibility in a parity-dependent matter. In conclusion, the present work proposes several novel marker associations and metabolic pathways contributing to genetic risk for HYK susceptibility.
Collapse
Affiliation(s)
- Ryan S Pralle
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nichol E Schultz
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Heather M White
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kent A Weigel
- Department of Dairy Science, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
49
|
Adablah JE, Wang Y, Donohue M, Roper MG. Profiling Glucose-Stimulated and M3 Receptor-Activated Insulin Secretion Dynamics from Islets of Langerhans Using an Extended-Lifetime Fluorescence Dye. Anal Chem 2020; 92:8464-8471. [PMID: 32429660 PMCID: PMC7304439 DOI: 10.1021/acs.analchem.0c01226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
![]()
Pulsatile insulin
from pancreatic islets is crucial for glucose
homeostasis, but the mechanism behind coordinated pulsatility is still
under investigation. One hypothesis suggests that cholinergic stimulation
of islets by pancreatic ganglia resets these endocrine units, producing
synchronization. Previously, it was shown that intracellular Ca2+ oscillations within islets can be entrained by pulses of
a cholinergic agonist, carbachol (CCh). Although these proxy measurements
of Ca2+ provided insight into the synchronization mechanism,
measurement of insulin output would be more direct evidence. To this
end, a fluorescence anisotropy competitive immunoassay for online
insulin detection from single and grouped islets in a microfluidic
system was developed using a piezoelectric pressure-driven fluid delivery
system and a squaraine rotaxane fluorophore, SeTau-647, as the fluorescent
label for insulin. Due to SeTau-647 having a longer lifetime and higher
brightness compared to the previously used Cy5 fluorophore, a 45%
increase in the anisotropy range was observed with enhanced signal-to-noise
ratio (S/N) of the measurements. This new system was tested by measuring
glucose-stimulated insulin secretion from single and groups of murine
and human islets. Distinct islet entrainment of groups of murine islets
by pulses of CCh was also observed, providing further evidence for
the hypothesis that pulsatile output from the ganglia can synchronize
islet behavior. We expect that this relatively straightforward, homogeneous
assay can be widely used for examining not only insulin secretion
but other secreted factors from different tissues.
Collapse
Affiliation(s)
- Joel E Adablah
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftain Way, Tallahassee, Florida 32306, United States
| | - Yao Wang
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftain Way, Tallahassee, Florida 32306, United States
| | - Matthew Donohue
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftain Way, Tallahassee, Florida 32306, United States
| | - Michael G Roper
- Department of Chemistry and Biochemistry, Florida State University, 95 Chieftain Way, Tallahassee, Florida 32306, United States
| |
Collapse
|
50
|
Non-coding RNA regulators of diabetic polyneuropathy. Neurosci Lett 2020; 731:135058. [PMID: 32454150 DOI: 10.1016/j.neulet.2020.135058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 02/08/2023]
Abstract
Diabetic polyneuropathy is a common and disturbing complication of diabetes mellitus, presenting patients and caregivers with a substantial disease burden. Emerging mechanisms which are underlying diabetes may provide novel pathways to understand diabetic polyneuropathy (DPN). Specifically, non-coding RNA molecules consisting of microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are implicated in the biological processes underlying DPN, and may link it to clinical spheres such as other metabolic and neural pathologies. Here, we elaborate on several candidate non-coding RNAs which may be associated with DPN via regulatory roles governing phenomena related to inflammatory, pain-provoking, and metabolic syndrome pathways. Specific examples include miRNAs such as miR-106a, -146a, -9, -29b, -466a, and -98; likewise, lncRNAs MIAT, PVT1, H19, MEG3, and MALAT1 are implicated, often co-affecting the involved pathways. Incorporating newly discovered regulators into what we know about specific clinical applications may highlight novel avenues for diagnosis, prevention, and intervention with DPN.
Collapse
|