1
|
Ahmad M, Wu S, Luo S, Shi W, Guo X, Cao Y, Perrimon N, He L. Dietary amino acids promote glucagon-like hormone release to generate global calcium waves in adipose tissues in Drosophila. Nat Commun 2025; 16:247. [PMID: 39747032 PMCID: PMC11696257 DOI: 10.1038/s41467-024-55371-y] [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: 05/28/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Propagation of intercellular calcium waves through tissues has been found to coordinate different multicellular responses. Nevertheless, our understanding of how calcium waves operate remains limited. In this study, we explore the real-time dynamics of intercellular calcium waves in Drosophila adipose tissues. We identify Adipokinetic Hormone (AKH), the fly functional homolog of glucagon, as the key factor driving Ca2+ activities in adipose tissue. We find that AKH, which is released into the hemolymph from the AKH-producing neurosecretory cells, stimulates calcium waves in the larval fat by a previously unrecognized gap-junction-independent mechanism to promote lipolysis. In the adult fat body, however, gap-junction-dependent intercellular calcium waves are triggered by a presumably uniformly diffused AKH. Additionally, we discover that amino acids activate the AKH-producing neurosecretory cells, leading to increased intracellular Ca2+ and AKH secretion. Altogether, we show that dietary amino acids regulate the AKH release from the AKH-producing neurosecretory cells in the brain, which subsequently stimulates gap-junction-independent intercellular calcium waves in adipose tissue, enhancing lipid metabolism.
Collapse
Affiliation(s)
- Muhammad Ahmad
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Shang Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shengyao Luo
- Yuanpei College, Peking University, Beijing, China
| | - Wenjia Shi
- Department of Applied Physics, Xi'an University of Technology, Xi'an, Shaanxi, China
| | - Xuan Guo
- Life Science Institute, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuansheng Cao
- Department of Physics, Tsinghua University, Beijing, China.
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| | - Li He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
2
|
Schuppelius B, Schüler R, Pivovarova-Ramich O, Hornemann S, Busjahn A, Machann J, Kruse M, Park SQ, Kabisch S, Csanalosi M, Ost AC, Pfeiffer AFH. Alterations in Glucagon Levels and the Glucagon-to-Insulin Ratio in Response to High Dietary Fat or Protein Intake in Healthy Lean Adult Twins: A Post Hoc Analysis. Nutrients 2024; 16:3905. [PMID: 39599691 PMCID: PMC11597242 DOI: 10.3390/nu16223905] [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: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Emerging data support evidence of the essential role of glucagon for lipid metabolism. However, data on the role of dietary fat intake for glucagon secretion is limited. This analysis investigated whether altering nutritional fat intake affects glucagon levels in healthy subjects. Methods: A total of 92 twins (age: 31 ± 14 years, BMI: 23 ± 3 kg/m2) consumed two 6-week diets: first a low-fat, high-carbohydrate diet (LFD) followed by an isocaloric high-fat, low-carbohydrate diet (HFD). In total, 24 twins (age: 39 ± 15 years, BMI: 24 ± 2 kg/m2) continued with a high-protein diet (HPD). Clinical investigations were performed after 6 weeks of the LFD, after 1 and 6 weeks of the HFD and after 6 weeks of the HPD. Results: The LFD caused a significant decrease in fasting glucagon (-27%, p < 0.001) compared to baseline. After 6 weeks of the HFD, glucagon increased (117%, p < 0.001 vs. LFD), while free fatty acids decreased. Six weeks of the HPD further increased glucagon levels (72%, p = 0.502 vs. HFD), although fasting amino acid levels remained constant. Fasting insulin and HOMA-IR moderately increased after one week of the HFD, while six weeks of the HPD significantly decreased both. The fasting glucagon-to-insulin ratio decreased during the LFD (p < 0.001) but increased after the HFD (p < 0.001) and even further increased after the HPD (p = 0.018). Liver fat, triglycerides and blood glucose did not increase during the HFD. The heritability of glucagon levels was 45% with the LFD. Conclusions: An HFD increases glucagon levels and the glucagon-to-insulin ratio under isocaloric conditions compared to an LFD in healthy lean subjects. This rise in glucagon may represent a metabolic response to prevent hepatic steatosis, as glucagon increases have been previously shown to induce hepatic fat oxidation.
Collapse
Affiliation(s)
- Bettina Schuppelius
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Rita Schüler
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Olga Pivovarova-Ramich
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Andreas Busjahn
- HealthTwiSt GmbH, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University of Tübingen, Geissweg 3, 72076 Tübingen, Germany
| | - Michael Kruse
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Soyoung Q. Park
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
- Department of Decision Neuroscience and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Neuroscience Research Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Stefan Kabisch
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| | - Marta Csanalosi
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Anne-Cathrin Ost
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| |
Collapse
|
3
|
Bomholt AB, Johansen CD, Galsgaard KD, Elmelund E, Winther-Sørensen M, Holst JJ, Wewer Albrechtsen NJ, Sørensen CM. Glucagon receptor activation contributes to the development of kidney injury. Am J Physiol Renal Physiol 2024; 327:F712-F724. [PMID: 39265079 PMCID: PMC11563637 DOI: 10.1152/ajprenal.00088.2024] [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: 03/18/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
The underlying causes of diabetic kidney disease are still largely unknown. New insights into the contributing causes of diabetic nephropathy are important to prevent this complication. Hyperglycemia and hypertension are some of the risk factors for diabetic nephropathy. However, the incidence of diabetic nephropathy is increasing despite efforts to normalize blood glucose levels and blood pressure. Therefore, other factors should be investigated as causes of diabetic nephropathy. We investigated whether long-term increased plasma levels of glucagon contribute to the development of pathophysiological changes in kidney function as seen in patients with diabetic nephropathy. Using mouse models of chronic activation and inactivation of glucagon receptor signaling, we investigated whether glucagon is involved in changes in renal function, renal structure, and transcriptional changes. We found several histopathological changes in the kidney, such as thickening of the parietal layer of Bowman's capsule, glomerular mesangial cell expansion, and significant albuminuria in the mice with activated glucagon receptor signaling. Opposite effects on mesangial area expansion and the development of albuminuria were demonstrated in mice with glucagon receptor inactivation. RNA sequencing data revealed that transcription of genes related to fatty acid metabolism, podocytes, Na+-K+-ATPase, and sodium/glucose transport was significantly changed in mice with activated glucagon receptor signaling. These data implicate that glucagon receptor signaling is involved in the development of kidney injury, as seen in type 2 diabetes, and that glucagon receptor is a potential therapeutic target in the treatment of diabetes. NEW & NOTEWORTHY This study suggests that the glucagon receptor is a potential therapeutic target in the treatment of diabetic kidney disease. We show, in mice, that long-term treatment with a glucagon analog showed not only pathophysiological changes and changes in renal function but also transcriptional changes in the kidneys, whereas opposite effects were demonstrated in mice with glucagon receptor inactivation. Therefore, the use of glucagon in a treatment regimen requires investigation of possible metabolic and renal abnormalities.
Collapse
Affiliation(s)
| | - Christian Dall Johansen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Elmelund
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital-Bispebjerg Hospital, Copenhagen, Denmark
| | | |
Collapse
|
4
|
Hendricks SA, Paul MJ, Subramaniam Y, Vijayam B. A collectanea of food insulinaemic index: 2023. Clin Nutr ESPEN 2024; 63:92-104. [PMID: 38941186 DOI: 10.1016/j.clnesp.2024.06.017] [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: 02/12/2024] [Revised: 04/28/2024] [Accepted: 06/11/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND AND AIMS To systematically update and publish the lnsulinaemic Index (II) value compilation of food/beverages. METHODS A literature search identified around 400 scholarly articles published between inception and December 2023. II values were pooled according to the selection criteria of at least 10 healthy, non-diabetic subjects with normal BMI. In addition, the II reported should have been derived from incremental area under the curve (iAUC) calculation of the insulin concentration over time. The reference food used from the pooled articles were either glucose or bread. RESULTS The II of 629 food/beverage items were found from 80 distinct articles. This is almost a five-fold increase in the number of entries from a previous compilation in 2011. Furthermore, these articles originated from 32 different countries, and were cleaved into 25 food categories. The II values ranged from 1 to 209. The highest overall recorded II was for a soy milk-based infant formula while the lowest was for both acacia fibre and gin. Upon clustering to single food, the infant formula retained the highest II while both acacia fibre and gin maintained the lowest recording. As for mixed meal, a potato dish served with a beverage recorded the highest II while a type of taco served with a sweetener, vegetable and fruit had the lowest II. Our minimum and maximum II data values replace the entries reported by previous compilations. CONCLUSION Acknowledging some limitations, these data would facilitate clinical usage of II for various applications in research, clinical nutrition, clinical medicine, diabetology and precision medicine. Future studies concerning II should investigate standardisation of reference food, including glucose and the test food portion. Although this collectanea adds up new food/beverages II values, priority should be given to populate this database.
Collapse
Affiliation(s)
| | | | - Yuganeswary Subramaniam
- Surgical Department, Hospital Besar Pulau Pinang, Jalan Residensi, 10990 Georgetown, Pulau Pinang, Malaysia
| | - Bhuwaneswaran Vijayam
- Newcastle University Medicine Malaysia (NUMed), Iskandar Puteri, 79200 Johor, Malaysia; Regenerative Medicine Working Group, Newcastle University Medicine Malaysia (NUMed), 79200 Iskandar Puteri, Johor, Malaysia.
| |
Collapse
|
5
|
Sisnande T, Brum FL, Matias DO, de Sá Ribeiro F, Moulin TB, Mohana-Borges R, de Magalhães MTQ, Lima LMTR. Spatially resolved distribution of pancreatic hormones proteoforms by MALDI-imaging mass spectrometry. Anal Biochem 2024; 692:115570. [PMID: 38763320 DOI: 10.1016/j.ab.2024.115570] [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: 11/07/2023] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
Zinc plays a crucial role both in the immune system and endocrine processes. Zinc restriction in the diet has been shown to lead to degeneration of the endocrine pancreas, resulting in hormonal imbalance within the β-cells. Proteostasismay vary depending on the stage of a pathophysiological process, which underscores the need for tools aimed at directly analyzing biological status. Among proteomics methods, MALDI-ToF-MS can serve as a rapid peptidomics tool for analyzing extracts or by histological imaging. Here we report the optimization of MALDI imaging mass spectrometry analysis of histological thin sections from mouse pancreas. This optimization enables the identification of the major islet peptide hormones as well as the major accumulated precursors and/or proteolytic products of peptide hormones. Cross-validation of the identified peptide hormones was performed by LC-ESI-MS from pancreatic islet extracts. Mice subjected to a zinc-restricted diet exhibited a relatively lower amount of peptide intermediates compared to the control group. These findings provide evidence for a complex modulation of proteostasis by micronutrients imbalance, a phenomenon directly accessed by MALDI-MSI.
Collapse
Affiliation(s)
- Tháyna Sisnande
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Programa de Pós-Graduação Em Química Biológica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Felipe Lopes Brum
- Laboratório de Biotecnologia e Bioengenharia Estrutural (LABGENEST), Instituto de Biofísica Carlos Chagas Filho (IBCCF), Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Daiane O Matias
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Programa de Pós-Graduação Em Química Biológica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Fernando de Sá Ribeiro
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Programa de Pós-Graduação Em Química Biológica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Thayana Beninatto Moulin
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural (LABGENEST), Instituto de Biofísica Carlos Chagas Filho (IBCCF), Rio de Janeiro, RJ, 21941-902, Brazil; Centro de Espectrometria de Massa de Biomoléculas (CEMBIO), Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Mariana T Q de Magalhães
- Laboratório de Biofísica de Macromoléculas (LBM), Instituto de Ciências Biomédicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil.
| | - Luís Maurício T R Lima
- Laboratório de Biotecnologia Farmacêutica (pbiotech), Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Programa de Pós-Graduação Em Química Biológica, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Programa de Pós-Graduação Em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
6
|
Félix-Martínez GJ, Osorio-Londoño D, Godínez-Fernández JR. Impact of oxygen and glucose availability on the viability and connectivity of islet cells: A computational study of reconstructed avascular human islets. PLoS Comput Biol 2024; 20:e1012357. [PMID: 39137218 PMCID: PMC11343470 DOI: 10.1371/journal.pcbi.1012357] [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: 05/14/2024] [Revised: 08/23/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
The experimental study and transplantation of pancreatic islets requires their isolation from the surrounding tissue, and therefore, from the vasculature. Under these conditions, avascular islets rely on the diffusion of peripheral oxygen and nutrients to comply with the requirements of islet cells while responding to changes in body glucose. As a complement to the experimental work, computational models have been widely used to estimate how avascular islets would be affected by the hypoxic conditions found both in culture and transplant sites. However, previous models have been based on simplified representations of pancreatic islets which has limited the reach of the simulations performed. Aiming to contribute with a more realistic model of avascular human islets, in this work we used architectures of human islets reconstructed from experimental data to simulate the availability of oxygen for α, β and δ-cells, emulating culture and transplant conditions at different glucose concentrations. The modeling approach proposed allowed us to quantitatively estimate how the loss of cells due to severe hypoxia would impact interactions between islet cells, ultimately segregating the islet into disconnected subnetworks. According to the simulations performed, islet encapsulation, by reducing the oxygen available within the islets, could severely compromise cell viability. Moreover, our model suggests that even without encapsulation, only microislets composed of less than 100 cells would remain viable in oxygenation conditions found in transplant sites. Overall, in this article we delineate a novel modeling methodology to simulate detailed avascular islets in experimental and transplant conditions with potential applications in the field of islet encapsulation.
Collapse
Affiliation(s)
- Gerardo J. Félix-Martínez
- Investigadoras e investigadores por México, Consejo Nacional de Humanidades, Ciencias y Tecnologías, México City, México
- Department of Electrical Engineering, Universidad Autónoma Metropolitana, Iztapalapa, México City, México
| | - Diana Osorio-Londoño
- Department of Electrical Engineering, Universidad Autónoma Metropolitana, Iztapalapa, México City, México
| | | |
Collapse
|
7
|
He L, Ahmad M, Wu S, Luo S, Shi W, Guo X, Cao Y, Perrimon N. Dietary Amino Acids Promote Glucagon-like Hormone Release to Generate Novel Calcium Waves in Adipose Tissues. RESEARCH SQUARE 2024:rs.3.rs-4493132. [PMID: 38947048 PMCID: PMC11213180 DOI: 10.21203/rs.3.rs-4493132/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Nutrient sensing and the subsequent metabolic responses are fundamental functions of animals, closely linked to diseases such as type 2 diabetes and various obesity-related morbidities. Among different metabolic regulatory signals, cytosolic Ca2+ plays pivotal roles in metabolic regulation, including glycolysis, gluconeogenesis, and lipolysis. Recently, intercellular calcium waves (ICWs), the propagation of Ca2+ signaling through tissues, have been found in different systems to coordinate multicellular responses. Nevertheless, our understanding of how ICWs are modulated and operate within living organisms remains limited. In this study, we explore the real-time dynamics, both in organ culture and free-behaving animals, of ICWs in Drosophila larval and adult adipose tissues. We identified Adipokinetic hormone (AKH), the fly functional homolog of mammalian glucagon, as the key factor driving Ca2+ activities in adipose tissue. Interestingly, we found that AKH, which is released in a pulsatile manner into the circulating hemolymph from the AKH-producing neurosecretory cells (APCs) in the brain, stimulates ICWs in the larval fat by a previously unrecognized gap-junction-independent mechanism to promote lipolysis. In the adult fat body, however, gap-junction-dependent random ICWs are triggered by a presumably uniformly diffused AKH. This highlights the stage-specific interplay of hormone secretion, extracellular diffusion, and intercellular communication in the regulation of Ca2+ dynamics. Additionally, we discovered that specific dietary amino acids activate the APCs, leading to increased intracellular Ca2+ and subsequent AKH secretion. Altogether, our findings identify that dietary amino acids regulate the release of AKH peptides from the APCs, which subsequently stimulates novel gap-junction-independent ICWs in adipose tissues, thereby enhancing lipid metabolism.
Collapse
Affiliation(s)
- Li He
- University of Science and Technology of China
| | | | - Shang Wu
- University of Science and Technology of China
| | | | | | | | | | | |
Collapse
|
8
|
Noguchi GM, Castillo VC, Donaldson CJ, Flisher MR, Momen AT, Saghatelian A, Huising MO. Urocortin 3 contributes to paracrine inhibition of islet alpha cells in mice. J Endocrinol 2024; 261:e240018. [PMID: 38593829 PMCID: PMC11095665 DOI: 10.1530/joe-24-0018] [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: 01/18/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
Pancreatic alpha cell activity and glucagon secretion lower as glucose levels increase. While part of the decrease is regulated by glucose itself, paracrine signaling by their neighboring beta and delta cells also plays an important role. Somatostatin from delta cells is an important local inhibitor of alpha cells at high glucose. Additionally, urocortin 3 (UCN3) is a hormone that is co-released from beta cells with insulin and acts locally to potentiate somatostatin secretion from delta cells. UCN3 thus inhibits insulin secretion via a negative feedback loop with delta cells, but its role with respect to alpha cells and glucagon secretion is not understood. We hypothesize that the somatostatin-driven glucagon inhibition at high glucose is regulated in part by UCN3 from beta cells. Here, we use a combination of live functional Ca2+ and cAMP imaging as well as direct glucagon secretion measurement, all from alpha cells in intact mouse islets, to determine the contributions of UCN3 to alpha cell behavior. Exogenous UCN3 treatment decreased alpha cell Ca2+ and cAMP levels and inhibited glucagon release. Blocking endogenous UCN3 signaling increased alpha cell Ca2+ by 26.8 ± 7.6%, but this did not result in increased glucagon release at high glucose. Furthermore, constitutive deletion of Ucn3 did not increase Ca2+ activity or glucagon secretion relative to controls. UCN3 is thus capable of inhibiting mouse alpha cells, but, given the subtle effects of endogenous UCN3 signaling on alpha cells, we propose that UCN3-driven somatostatin may serve to regulate local paracrine glucagon levels in the islet instead of inhibiting gross systemic glucagon release.
Collapse
Affiliation(s)
- Glyn M. Noguchi
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, USA
| | - Vincent C. Castillo
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, USA
| | | | - Marcus R. Flisher
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, USA
| | - Ariana T. Momen
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, USA
| | | | - Mark O. Huising
- Department of Neurobiology, Physiology & Behavior, University of California Davis, Davis, CA, USA
- Department of Physiology & Membrane Biology, University of California Davis, Davis, CA, USA
| |
Collapse
|
9
|
Kistkins S, Moser O, Ankudovičs V, Blizņuks D, Mihailovs T, Lobanovs S, Sourij H, Pfeiffer AFH, Pīrāgs V. From classical dualistic antagonism to hormone synergy: potential of overlapping action of glucagon, insulin and GLP-1 for the treatment of diabesity. Endocr Connect 2024; 13:e230529. [PMID: 38579770 PMCID: PMC11046332 DOI: 10.1530/ec-23-0529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 04/03/2024] [Indexed: 04/07/2024]
Abstract
The increasing prevalence of 'diabesity', a combination of type 2 diabetes and obesity, poses a significant global health challenge. Unhealthy lifestyle factors, including poor diet, sedentary behaviour, and high stress levels, combined with genetic and epigenetic factors, contribute to the diabesity epidemic. Diabesity leads to various significant complications such as cardiovascular diseases, stroke, and certain cancers. Incretin-based therapies, such as GLP-1 receptor agonists and dual hormone therapies, have shown promising results in improving glycaemic control and inducing weight loss. However, these therapies also come with certain disadvantages, including potential withdrawal effects. This review aims to provide insights into the cross-interactions of insulin, glucagon, and GLP-1, revealing the complex hormonal dynamics during fasting and postprandial states, impacting glucose homeostasis, energy expenditure, and other metabolic functions. Understanding these hormonal interactions may offer novel hypotheses in the development of 'anti-diabesity' treatment strategies. The article also explores the question of the antagonism of insulin and glucagon, providing insights into the potential synergy and hormonal overlaps between these hormones.
Collapse
Affiliation(s)
| | - Othmar Moser
- Division of Exercise Physiology and Metabolism, Institute of Sport Science, University of Bayreuth, Bayreuth, Germany
| | | | - Dmitrijs Blizņuks
- Institute of Smart Computing Technologies, Riga Technical University, Riga, Latvia
| | - Timurs Mihailovs
- Institute of Smart Computing Technologies, Riga Technical University, Riga, Latvia
| | | | - Harald Sourij
- Trials Unit for Interdisciplinary Metabolic Medicine, Division of Endocrinology and Diabetolgoy, Medical University of Graz, Graz, Austria
| | - Andreas F H Pfeiffer
- Department of Endocrinology and Metabolic Medicine, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm, Berlin, Germany
| | - Valdis Pīrāgs
- Pauls Stradiņš Clinical University Hospital, Riga, Latvia
- Faculty of Medicine, University of Latvia, Riga, Latvia
| |
Collapse
|
10
|
Chang L, Ma X, Yuan M, Ding L, Gu Y, Liu L, Li Y, Shu H, Liu M, He Q. Lowest Glucagon/Highest C-Peptide in Oral Glucose Tolerance Test: Clinical Utility in Monitoring Glucose Control in Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2024; 17:1769-1780. [PMID: 38645659 PMCID: PMC11032662 DOI: 10.2147/dmso.s459392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/10/2024] [Indexed: 04/23/2024] Open
Abstract
Purpose Understanding factors that influence blood glucose levels in patients with type 2 diabetes mellitus (T2DM) is crucial for managing hyperglycemia. Currently, there is no standardized interpretation method for glucagon levels in oral glucose tolerance test (OGTT). This study aims to assess the relationship between the lowest glucagon/highest C-peptide ratio (Lglc/Hcp) in OGTT and glucose control levels in T2DM. Patients and Methods Clinical data from 120 patients with T2DM were examined to compare the correlations of Lglc/Hcp and other pancreatic islet function-associated indices with fasting blood glucose (G0), glucose at 120 minutes in OGTT (G120), hemoglobin A1c (HbA1c), and the area under the glucose curve in OGTT (AUCglu). Additionally, the study investigated difference in Lglc/Hcp between patient groups based on the highest blood glucose levels (Hglu) in OGTT (Hglu ≥ 16.7 mmol/L vs Hglu < 16.7 mmol/L). Results The generalized linear model suggested that Lglc/Hcp significantly correlated with G0 (B = 0.85, P < 0.001), G120(B = 1.46, P < 0.001), HbA1c (B = 0.67, P < 0.001), and AUCglu (B = 3.46, P < 0.001). This correlation surpassed C-peptide and glucagon-related parameters, even after adjusting for confounding factors. Furthermore, Lglc/Hcp was notably higher in patients with Hglu ≥ 16.7 mmol/L compared to those with Hglu < 16.7 mmol/L (Z = -3.71, p < 0.001). Conclusion Lglc/Hcp in OGTT closely relates to blood glucose control in patients with T2DM, potentially reflecting the overall pancreatic islet function in regulating glucose levels. Moreover, inhibiting glucagon secretion may be a crucial consideration for patients requiring insulin treatment.
Collapse
Affiliation(s)
- Lina Chang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Xiaohui Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Menghua Yuan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Yian Gu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Lili Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Yan Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Hua Shu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
11
|
Ahmad M, Wu S, Guo X, Perrimon N, He L. Sensing of dietary amino acids and regulation of calcium dynamics in adipose tissues through Adipokinetic hormone in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583442. [PMID: 38496667 PMCID: PMC10942355 DOI: 10.1101/2024.03.04.583442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Nutrient sensing and the subsequent metabolic responses are fundamental functions of animals, closely linked to diseases such as type 2 diabetes and various obesity-related diseases. Drosophila melanogaster has emerged as an excellent model for investigating metabolism and its associated disorders. In this study, we used live-cell imaging to demonstrate that the fly functional homolog of mammalian glucagon, Adipokinetic hormone (AKH), secreted from AKH hormone-producing cells (APCs) in the corpora cardiaca, stimulates intracellular Ca 2+ waves in the larval fat body/adipose tissue to promote lipid metabolism. Further, we show that specific dietary amino acids activate the APCs, leading to increased intracellular Ca 2+ and subsequent AKH secretion. Finally, a comparison of Ca 2+ dynamics in larval and adult fat bodies revealed different mechanisms of regulation, highlighting the interplay of pulses of AKH secretion, extracellular diffusion of the hormone, and intercellular communication through gap junctions. Our study underscores the suitability of Drosophila as a powerful model for exploring real-time nutrient sensing and inter-organ communication dynamics.
Collapse
|
12
|
Zhang J, Zheng Y, Martens L, Pfeiffer AFH. The Regulation and Secretion of Glucagon in Response to Nutrient Composition: Unraveling Their Intricate Mechanisms. Nutrients 2023; 15:3913. [PMID: 37764697 PMCID: PMC10536047 DOI: 10.3390/nu15183913] [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: 08/01/2023] [Revised: 08/31/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Glucagon was initially regarded as a hyperglycemic substance; however, recent research has revealed its broader role in metabolism, encompassing effects on glucose, amino acids (AAs), and lipid metabolism. Notably, the interplay of glucagon with nutrient intake, particularly of AAs, and non-nutrient components is central to its secretion. Fasting and postprandial hyperglucagonemia have long been linked to the development and progression of type 2 diabetes (T2DM). However, recent studies have brought to light the positive impact of glucagon agonists on lipid metabolism and energy homeostasis. This review explores the multifaceted actions of glucagon, focusing on its regulation, signaling pathways, and effects on glucose, AAs, and lipid metabolism. The interplay between glucagon and other hormones, including insulin and incretins, is examined to provide a mechanistic understanding of its functions. Notably, the liver-α-cell axis, which involves glucagon and amino acids, emerges as a critical aspect of metabolic regulation. The dysregulation of glucagon secretion and its impact on conditions such as T2DM are discussed. The review highlights the potential therapeutic applications of targeting the glucagon pathway in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Jiudan Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| | - Yang Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, China;
| | - Lisa Martens
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
- Nutritional Science, University of Potsdam, 14469 Potsdam, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (L.M.); (A.F.H.P.)
| |
Collapse
|
13
|
Bang J, Lee SA, Koh G, Yoo S. Association of Glucagon to Insulin Ratio and Metabolic Syndrome in Patients with Type 2 Diabetes. J Clin Med 2023; 12:5806. [PMID: 37762748 PMCID: PMC10531641 DOI: 10.3390/jcm12185806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/06/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
There is a growing interest in the role of glucagon in type 2 diabetes mellitus (T2DM). Glucagon and insulin regulate glucose and lipid metabolism. Metabolic syndrome is an important risk factor for cardiovascular disease in patients with T2DM. We investigated the association between glucagon to insulin ratio and metabolic syndrome in patients with T2DM. This is a cross-sectional study involving 317 people with type 2 diabetes. Glucagon and insulin levels were measured in a fasted state and 30 min after ingesting a standard mixed meal. The Criteria of the International Diabetes Federation defined metabolic syndrome. Two hundred nineteen (69%) of the subjects had metabolic syndrome. The fasting glucagon to insulin ratio was significantly lower in patients with metabolic syndrome (14.0 ± 9.7 vs. 17.3 ± 10.3, p < 0.05). The fasting glucagon to insulin ratio was significantly lowered as the number of metabolic syndrome components increased. In hierarchical logistic regression analysis, the fasting glucagon to insulin ratio significantly contributed to metabolic syndrome even after adjusting for other covariates. The fasting glucagon to insulin ratio is inversely associated with metabolic syndrome in patients with type 2 diabetes. This suggests that glucagon-targeted therapeutics may reduce cardiovascular risk by improving metabolic syndrome.
Collapse
Affiliation(s)
- Jisun Bang
- Department of Internal Medicine, Jeju National University Hospital, Jeju 63241, Republic of Korea
| | - Sang Ah Lee
- Department of Internal Medicine, Jeju National University Hospital, Jeju 63241, Republic of Korea
- Department of Internal Medicine, Jeju National University College of Medicine, Jeju 63241, Republic of Korea
| | - Gwanpyo Koh
- Department of Internal Medicine, Jeju National University Hospital, Jeju 63241, Republic of Korea
- Department of Internal Medicine, Jeju National University College of Medicine, Jeju 63241, Republic of Korea
| | - Soyeon Yoo
- Department of Internal Medicine, Jeju National University Hospital, Jeju 63241, Republic of Korea
- Department of Internal Medicine, Jeju National University College of Medicine, Jeju 63241, Republic of Korea
| |
Collapse
|
14
|
St. Clair JR, Westacott MJ, Miranda J, Farnsworth NL, Kravets V, Schleicher WE, Dwulet JM, Levitt CH, Heintz A, Ludin NWF, Benninger RKP. Restoring connexin-36 function in diabetogenic environments precludes mouse and human islet dysfunction. J Physiol 2023; 601:4053-4072. [PMID: 37578890 PMCID: PMC10508056 DOI: 10.1113/jp282114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/27/2023] [Indexed: 08/16/2023] Open
Abstract
The secretion of insulin from β-cells in the islet of Langerhans is governed by a series of metabolic and electrical events, which can fail during the progression of type 2 diabetes (T2D). β-cells are electrically coupled via connexin-36 (Cx36) gap junction channels, which coordinates the pulsatile dynamics of [Ca2+ ] and insulin release across the islet. Factors such as pro-inflammatory cytokines and free fatty acids disrupt gap junction coupling under in vitro conditions. Here we test whether gap junction coupling and coordinated [Ca2+ ] dynamics are disrupted in T2D, and whether recovery of gap junction coupling can recover islet function. We examine islets from donors with T2D, from db/db mice, and islets treated with pro-inflammatory cytokines (TNF-α, IL-1β, IFN-ɣ) or free fatty acids (palmitate). We modulate gap junction coupling using Cx36 over-expression or pharmacological activation via modafinil. We also develop a peptide mimetic (S293) of the c-terminal regulatory site of Cx36 designed to compete against its phosphorylation. Cx36 gap junction permeability and [Ca2+ ] dynamics were disrupted in islets from both human donors with T2D and db/db mice, and in islets treated with pro-inflammatory cytokines or palmitate. Cx36 over-expression, modafinil treatment and S293 peptide all enhanced Cx36 gap junction coupling and protected against declines in coordinated [Ca2+ ] dynamics. Cx36 over-expression and S293 peptide also reduced apoptosis induced by pro-inflammatory cytokines. Critically, S293 peptide rescued gap junction coupling and [Ca2+ ] dynamics in islets from both db/db mice and a sub-set of T2D donors. Thus, recovering or enhancing Cx36 gap junction coupling can improve islet function in diabetes. KEY POINTS: Connexin-36 (Cx36) gap junction permeability and associated coordination of [Ca2+ ] dynamics is diminished in human type 2 diabetes (T2D) and mouse models of T2D. Enhancing Cx36 gap junction permeability protects against disruptions to the coordination of [Ca2+ ] dynamics. A novel peptide mimetic of the Cx36 c-terminal regulatory region protects against declines in Cx36 gap junction permeability. Pharmacological elevation in Cx36 or Cx36 peptide mimetic recovers [Ca2+ ] dynamics and glucose-stimulated insulin secretion in human T2D and mouse models of T2D.
Collapse
Affiliation(s)
- Joshua R St. Clair
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Matthew J Westacott
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Jose Miranda
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Nikki L Farnsworth
- Barbara Davis Center for Diabetes, University of Colorado
Denver | Anschutz Medical Campus, Aurora, CO
| | - Vira Kravets
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Wolfgang E Schleicher
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - JaeAnn M Dwulet
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Claire H Levitt
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Audrey Heintz
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Nurin WF Ludin
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
| | - Richard KP Benninger
- Department of Bioengineering, University of Colorado Denver
| Anschutz Medical Campus, Aurora, CO
- Barbara Davis Center for Diabetes, University of Colorado
Denver | Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
15
|
Adams MT, Waters BJ, Nimkulrat SD, Blum B. Disrupted glucose homeostasis and glucagon and insulin secretion defects in Robo βKO mice. FASEB J 2023; 37:e23106. [PMID: 37498234 PMCID: PMC10436995 DOI: 10.1096/fj.202200705rr] [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: 05/17/2022] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
The axon guidance proteins, Roundabout (Robo) receptors play a critical role in morphogenesis of the islets of Langerhans. Mice with a β cell-selective deletion of Robo (Robo βKO), show severely disrupted spatial architecture of their islets, without defects in β cell differentiation or maturity. We have recently shown that Robo βKO mice have reduced synchronous glucose-stimulated β cell calcium oscillations in their islets in vivo, likely disrupting their pulsatile insulin secretion. Here, we analyze whole-body metabolic regulation in Robo βKO mice. We show that Robo βKO mice have mild defects in glucose homeostasis, and altered glucagon and insulin secretion. However, we did not observe any severe whole-body glucoregulatory phenotype following the disruption of islet architecture in Robo βKO. Our data suggest that islet architecture plays only a mild role in overall glucoregulation.
Collapse
Affiliation(s)
- Melissa T. Adams
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Bayley J. Waters
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Sutichot D. Nimkulrat
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Barak Blum
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
16
|
Zhang J, Schäfer SM, Kabisch S, Csanalosi M, Schuppelius B, Kemper M, Markova M, Meyer NMT, Pivovarova-Ramich O, Keyhani-Nejad F, Rohn S, Pfeiffer AFH. Implication of sugar, protein and incretins in excessive glucagon secretion in type 2 diabetes after mixed meals. Clin Nutr 2023; 42:467-476. [PMID: 36857956 DOI: 10.1016/j.clnu.2023.02.011] [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: 07/04/2022] [Revised: 01/10/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023]
Abstract
AIMS Amino acids powerfully release glucagon but their contribution to postprandial hyperglucagonemia in type 2 diabetes remains unclear. Exogenously applied GIP stimulates, while GLP-1 inhibits, glucagon secretion in humans. However, their role in mixed meals is unclear, which we therefore characterized. METHODS In three experiments, participants with type 2 diabetes and obese controls randomly received different loads of sugars and/or proteins. In the first experiment, participants ingested the rapidly cleaved saccharose (SAC) or slowly cleaved isomaltulose (ISO) which is known to elicit opposite profiles of GIP and GLP-1 secretion. In the second one participants received test meals which contained saccharose or isomaltulose in combination with milk protein. The third set of participants underwent randomized oral protein tests with whey protein or casein. Incretins, glucagon, C-peptide, and insulin were profiled by specific immunological assays. RESULTS 50 g of the sugars alone suppressed glucagon in controls but slightly less in type 2 diabetes patients. Participants with type 2 diabetes showed excessive glucagon responses within 15 min and lasting over 3 h, while the obese controls showed small initial and delayed greater glucagon responses to mixed meals. The release of GIP was significantly faster and greater with SAC compared to ISO, while GLP-1 showed an inverse pattern. The glucagon responses to whey or casein were only moderately increased in type 2 diabetes patients without a left shift of the dose response curve. CONCLUSIONS The rapid hypersecretion of glucagon after mixed meals in type 2 diabetes patients compared to controls is unaffected by endogenous incretins. The defective suppression of glucagon by glucose combined with hypersecretion to protein is required for the exaggerated response. CLINICAL TRIALS NUMBERS NCT03806920, NCT02219295, NCT04564391.
Collapse
Affiliation(s)
- Jiudan Zhang
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Sylva Mareike Schäfer
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; Institute of Nutritional Science, Justus-Liebig University of Giessen, Giessen, Germany
| | - Stefan Kabisch
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Marta Csanalosi
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Bettina Schuppelius
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Margrit Kemper
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Mariya Markova
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Nina Marie Tosca Meyer
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Olga Pivovarova-Ramich
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany; Reseach Group Molecular Nutritional Medicine, Dept. of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbrücke, 14558, Nuthetal, Germany
| | - Farnaz Keyhani-Nejad
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Sascha Rohn
- Institute of Food Chemistry, Hamburg School of Food Science, University of Hamburg, Grindelallee 117, 20146, Hamburg, Germany; Institute of Food Technology and Food Chemistry, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355, Berlin, Germany
| | - Andreas F H Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany; German Center for Diabetes Research (Deutsches Zentrum Für Diabetesforschung e.V.), Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| |
Collapse
|
17
|
Xia Z, You W, Li Y, Li F, Hao S, Sun Y, Li N, Lin L, Dou J, Su X, Zhai Q, Zuo Y, Zhang Y, Gaisano HY, Zheng D, He Y, Jiang J. Association between residual islet beta-cell function and achieving the target of time in range in inpatients with type 2 diabetes undergoing antidiabetic treatment: An observation study. Diabetes Obes Metab 2023; 25:1714-1722. [PMID: 36811214 DOI: 10.1111/dom.15026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
AIM To assess whether the beta-cell function of inpatients undergoing antidiabetic treatment influences achieving time in range (TIR) and time above range (TAR) targets. MATERIALS AND METHODS This cross-sectional study included 180 inpatients with type 2 diabetes. TIR and TAR were assessed by a continuous glucose monitoring system, with target achievement defined as TIR more than 70% and TAR less than 25%. Beta-cell function was assessed by the insulin secretion-sensitivity index-2 (ISSI2). RESULTS Following antidiabetic treatment, logistic regression analysis showed that lower ISSI2 was associated with a decreased number of inpatients achieving TIR (OR = 3.10, 95% CI: 1.19-8.06) and TAR (OR = 3.40, 95% CI: 1.35-8.55) targets after adjusting for potential confounders. Similar associations still existed in those participants treated with insulin secretagogues (TIR: OR = 2.91, 95% CI: 0.90-9.36, P = .07; TAR, OR = 3.14, 95% CI: 1.01-9.80) or adequate insulin therapy (TIR: OR = 2.84, 95% CI: 0.91-8.81, P = .07; TAR, OR = 3.24, 95% CI: 1.08-9.67). Furthermore, receiver operating characteristic curves showed that the diagnostic value of the ISSI2 for achieving TIR and TAR targets was 0.73 (95% CI: 0.66-0.80) and 0.71 (95% CI: 0.63-0.79), respectively. CONCLUSIONS Beta-cell function was associated with achieving TIR and TAR targets. Stimulating insulin secretion or exogenous insulin treatment could not overcome the disadvantage of lower beta-cell function on glycaemic control.
Collapse
Affiliation(s)
- Zhang Xia
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Wenjun You
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
| | - Yuhao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Feng Li
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
- Institute for Chronic Disease Management, Jining No.1 People's Hospital, Jining, China
| | - Shuai Hao
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
| | - Yihan Sun
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
| | - Na Li
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
| | - Lu Lin
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jingtao Dou
- Department of Endocrinology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xin Su
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Qi Zhai
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yingting Zuo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yibo Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Herbert Y Gaisano
- Departments of Medication and Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Deqiang Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Yan He
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Jiajia Jiang
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
- Institute for Chronic Disease Management, Jining No.1 People's Hospital, Jining, China
| |
Collapse
|
18
|
Laurenti MC, Arora P, Dalla Man C, Andrews JC, Rizza RA, Matveyenko A, Bailey KR, Cobelli C, Vella A. The relationship between insulin and glucagon concentrations in non-diabetic humans. Physiol Rep 2022; 10:e15380. [PMID: 35822422 PMCID: PMC9277417 DOI: 10.14814/phy2.15380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 06/02/2023] Open
Abstract
Abnormal postprandial suppression of glucagon in Type 2 diabetes (T2DM) has been attributed to impaired insulin secretion. Prior work suggests that insulin and glucagon show an inverse coordinated relationship. However, dysregulation of α-cell function in prediabetes occurs early and independently of changes in β-cells, which suggests insulin having a less significant role on glucagon control. We therefore, sought to examine whether hepatic vein hormone concentrations provide evidence to further support the modulation of glucagon secretion by insulin. As part of a series of experiments to measure the effect of diabetes-associated genetic variation in TCF7L2 on islet cell function, hepatic vein insulin and glucagon concentrations were measured at 2-minute intervals during fasting and a hyperglycemic clamp. The experiment was performed on 29 nondiabetic subjects (age = 46 ± 2 years, BMI 28 ± 1 Kg/m2 ) and enabled post-hoc analysis, using Cross-Correlation and Cross-Approximate Entropy (Cross-ApEn) to evaluate the interaction of insulin and glucose. Mean insulin concentrations rose from fasting (33 ± 4 vs. 146 ± 12 pmol/L, p < 0.01) while glucagon was suppressed (96 ± 8 vs. 62 ± 5 ng/L, p < 0.01) during the clamp. Cross-ApEn was used to measure pattern reproducibility in the two hormones using glucagon as control mechanism (0.78 ± 0.03 vs. 0.76 ± 0.03, fasting vs. hyperglycemia) and using insulin as a control mechanism (0.78 ± 0.02 vs. 0.76 ± 0.03, fasting vs. hyperglycemia). Values did not differ between the two scenarios. Cross-correlation analysis demonstrated a small in-phase coordination between insulin and glucagon concentrations during fasting, which inverted during hyperglycemia. This data suggests that the interaction between the two hormones is not driven by either. On a minute-to-minute basis, direct control and secretion of glucagon is not mediated (or restrained) by insulin.
Collapse
Affiliation(s)
- Marcello C. Laurenti
- Division of Endocrinology, Diabetes & MetabolismEndocrine Research Unit, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical SciencesRochesterMinnesotaUSA
| | - Praveer Arora
- Division of Endocrinology, Diabetes & MetabolismEndocrine Research Unit, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
| | - Chiara Dalla Man
- Department of Information EngineeringUniversity of PadovaPadovaItaly
| | - James C. Andrews
- Vascular and Interventional Radiology, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
| | - Robert A. Rizza
- Division of Endocrinology, Diabetes & MetabolismEndocrine Research Unit, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
| | - Aleksey Matveyenko
- Division of Endocrinology, Diabetes & MetabolismEndocrine Research Unit, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
| | - Kent R. Bailey
- Division of Biomedical Statistics and Informatics, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
| | - Claudio Cobelli
- Department of Woman and Child's HealthUniversity of PadovaPadovaItaly
| | - Adrian Vella
- Division of Endocrinology, Diabetes & MetabolismEndocrine Research Unit, Mayo Clinic, College of Medicine and ScienceRochesterMinnesotaUSA
| |
Collapse
|
19
|
Ren H, Li Y, Han C, Yu Y, Shi B, Peng X, Zhang T, Wu S, Yang X, Kim S, Chen L, Tang C. Pancreatic α and β cells are globally phase-locked. Nat Commun 2022; 13:3721. [PMID: 35764654 PMCID: PMC9240067 DOI: 10.1038/s41467-022-31373-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 06/15/2022] [Indexed: 11/25/2022] Open
Abstract
The Ca2+ modulated pulsatile glucagon and insulin secretions by pancreatic α and β cells play a crucial role in glucose homeostasis. However, how α and β cells coordinate to produce various Ca2+ oscillation patterns is still elusive. Using a microfluidic device and transgenic mice, we recorded Ca2+ signals from islet α and β cells, and observed heterogeneous Ca2+ oscillation patterns intrinsic to each islet. After a brief period of glucose stimulation, α and β cells’ oscillations were globally phase-locked. While the activation of α cells displayed a fixed time delay of ~20 s to that of β cells, β cells activated with a tunable period. Moreover, islet α cell number correlated with oscillation frequency. We built a mathematical model of islet Ca2+ oscillation incorporating paracrine interactions, which quantitatively agreed with the experimental data. Our study highlights the importance of cell-cell interaction in generating stable but tunable islet oscillation patterns. The Ca2+ modulated pulsatile glucagon and insulin secretions by pancreatic α and β cells are critical in glucose homeostasis. Here the authors show that the Ca2+ oscillations of α and β cells are phase-locked, and that the oscillation pattern is tuned by paracrine interactions between α and β cells.
Collapse
Affiliation(s)
- Huixia Ren
- Center for Quantitative Biology, Peking University, Beijing, 100871, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yanjun Li
- Center for Quantitative Biology, Peking University, Beijing, 100871, China.,Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, 100871, China
| | - Chengsheng Han
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, 100871, China
| | - Yi Yu
- Center for Quantitative Biology, Peking University, Beijing, 100871, China
| | - Bowen Shi
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Xiaohong Peng
- Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, 100871, China
| | - Tianming Zhang
- Yuanpei College, Peking University, Beijing, 100871, China
| | - Shufang Wu
- Center for Quantitative Biology, Peking University, Beijing, 100871, China
| | - Xiaojing Yang
- Center for Quantitative Biology, Peking University, Beijing, 100871, China.,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Sneppen Kim
- Niels Bohr Institute, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Liangyi Chen
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China. .,Institute of Molecular Medicine, School of Future Technology, National Biomedical Imaging Center, Peking University, Beijing, 100871, China.
| | - Chao Tang
- Center for Quantitative Biology, Peking University, Beijing, 100871, China. .,Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
20
|
Zhang J, Pivovarova-Ramich O, Kabisch S, Markova M, Hornemann S, Sucher S, Rohn S, Machann J, Pfeiffer AFH. High Protein Diets Improve Liver Fat and Insulin Sensitivity by Prandial but Not Fasting Glucagon Secretion in Type 2 Diabetes. Front Nutr 2022; 9:808346. [PMID: 35662921 PMCID: PMC9160603 DOI: 10.3389/fnut.2022.808346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Glucagon (GCGN) plays a key role in glucose and amino acid (AA) metabolism by increasing hepatic glucose output. AA strongly stimulate GCGN secretion which regulates hepatic AA degradation by ureagenesis. Although increased fasting GCGN levels cause hyperglycemia GCGN has beneficial actions by stimulating hepatic lipolysis and improving insulin sensitivity through alanine induced activation of AMPK. Indeed, stimulating prandial GCGN secretion by isocaloric high protein diets (HPDs) strongly reduces intrahepatic lipids (IHLs) and improves glucose metabolism in type 2 diabetes mellitus (T2DM). Therefore, the role of GCGN and circulating AAs in metabolic improvements in 31 patients with T2DM consuming HPD was investigated. Six weeks HPD strongly coordinated GCGN and AA levels with IHL and insulin sensitivity as shown by significant correlations compared to baseline. Reduction of IHL during the intervention by 42% significantly improved insulin sensitivity [homeostatic model assessment for insulin resistance (HOMA-IR) or hyperinsulinemic euglycemic clamps] but not fasting GCGN or AA levels. By contrast, GCGN secretion in mixed meal tolerance tests (MMTTs) decreased depending on IHL reduction together with a selective reduction of GCGN-regulated alanine levels indicating greater GCGN sensitivity. HPD aligned glucose metabolism with GCGN actions. Meal stimulated, but not fasting GCGN, was related to reduced liver fat and improved insulin sensitivity. This supports the concept of GCGN-induced hepatic lipolysis and alanine- and ureagenesis-induced activation of AMPK by HPD.
Collapse
Affiliation(s)
- Jiudan Zhang
- Department of Endocrinology, Diabetes and Nutrition, Charité – Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Jiudan Zhang,
| | - Olga Pivovarova-Ramich
- Department of Endocrinology, Diabetes and Nutrition, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Potsdam, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
| | - Stefan Kabisch
- Department of Endocrinology, Diabetes and Nutrition, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Potsdam, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
| | - Mariya Markova
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Potsdam, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Potsdam, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
| | - Stephanie Sucher
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Potsdam, Germany
| | - Sascha Rohn
- Hamburg School of Food Science, Institute of Food Chemistry, University of Hamburg, Hamburg, Germany
- Faculty of Process Sciences, Institute of Food Technology and Food Chemistry, Technical University of Berlin, Berlin, Germany
| | - Jürgen Machann
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital, Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Deutsches Zentrum für Diabetesforschung (DZD), Neuherberg, Germany
- Andreas F. H. Pfeiffer,
| |
Collapse
|
21
|
Wu XD, Huang S, Shi Y, Shen Y, Tu WC, Leng Y, Zhao QS. Design, synthesis and structural-activity relationship studies of phanginin A derivatives for regulating SIK1-cAMP/CREB signaling to suppress hepatic gluconeogenesis. Eur J Med Chem 2022; 232:114171. [DOI: 10.1016/j.ejmech.2022.114171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/24/2022] [Accepted: 01/30/2022] [Indexed: 11/30/2022]
|
22
|
Chai S, Zhang R, Zhang Y, Carr RD, Zheng Y, Rajpathak S, Ji L. Effect of dipeptidyl peptidase-4 inhibitors on postprandial glucagon level in patients with type 2 diabetes mellitus: A systemic review and meta-analysis. Front Endocrinol (Lausanne) 2022; 13:994944. [PMID: 36313782 PMCID: PMC9597445 DOI: 10.3389/fendo.2022.994944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Hyperglucagonemia occurs in the pathogenesis of type 2 diabetes mellitus (T2DM). In this meta-analysis, we summarized the effects of DPP4 inhibitors on glucagon levels in patients with T2DM. MATERIALS AND METHODS Randomized controlled trials (RCTs) comparing the influence of DPP4 inhibitors on circulating glucagon levels with placebo or other oral antidiabetic drugs (OADs) in patients with T2DM were identified by searches of Medline (PubMed), Embase (Ovid), and CENTER (Cochrane Library). Only studies reporting changes in glucagon level presented as total area under the curve (AUCglucagon) using a meal or oral glucose tolerance test were included. Results were combined using a random-effects model that incorporated potential heterogeneity among the included studies. RESULTS A total of 36 RCTs with moderate to high quality were included. Overall, the numbers of T2DM patients included for the meta-analyses comparing DPP4 inhibitors with placebo and other OADs were 4266 and 1652, respectively. Compared to placebo, DPP4 inhibitors significantly reduced circulating glucagon levels (standard mean difference [SMD]: -0.32, 95% CI: -0.40 to -0.24, P<0.001; I2 = 28%). Analysis of subgroups revealed that study characteristics had no significant effect on results, such as study design (parallel group or crossover), number of patients, mean patient age, proportion of men, baseline HbA1c, duration of diabetes, background therapy, treatment duration, or methods for glucagon measurement (all P for subgroup differences >0.05). Moreover, DPP4 inhibitors significantly reduced glucagon levels compared to other OADs (SMD: -0.35, 95% CI: -0.53 to -0.16, P<0.001; I2 = 66%), and the reduction in glucagon was greater in comparison with insulin secretagogues than in comparison with non-insulin secretagogues (P for subgroup difference =0.03). SYSTEMATIC REVIEW REGISTRATION https://inplasy.com/, identifier INPLASY202280104. CONCLUSIONS DPP4 inhibitors are effective at reducing the circulating postprandial glucagon level in T2DM patients.
Collapse
Affiliation(s)
- Shangyu Chai
- Merck Research Laboratories (MRL) Global Medical Affairs, Merck Sharp & Dohme (MSD) China, Shanghai, China
| | - Ruya Zhang
- Merck Research Laboratories (MRL) Global Medical Affairs, Merck Sharp & Dohme (MSD) China, Shanghai, China
| | - Ye Zhang
- Merck Research Laboratories (MRL) Global Medical Affairs, Merck Sharp & Dohme (MSD) China, Shanghai, China
| | - Richard David Carr
- Hatter Cardiovascular Institute, University College London, UK and Ulster University, Coleraine, United Kingdom
| | - Yiman Zheng
- Merck Research Laboratories (MRL) Global Medical Affairs, Merck Sharp & Dohme (MSD) China, Shanghai, China
| | - Swapnil Rajpathak
- Merck Research Laboratories, Merck & Co., Inc., Rahway, NJ, United States
| | - Linong Ji
- Department of Endocrinology, People’s Hospital of Peking University, Beijing, China
- *Correspondence: Linong Ji,
| |
Collapse
|
23
|
Continuous variable responses and signal gating form kinetic bases for pulsatile insulin signaling and emergence of resistance. Proc Natl Acad Sci U S A 2021; 118:2102560118. [PMID: 34615716 PMCID: PMC8522282 DOI: 10.1073/pnas.2102560118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 12/16/2022] Open
Abstract
Evolutionarily conserved insulin signaling is central to nutrient sensing, storage, and utilization across tissues. Dysfunctional insulin signaling is associated with metabolic disorders, cancer, and aging. Hence, the pathway components have emerged as key targets for pharmacological interventions in addition to insulin administration itself. Despite this, activation–inactivation dynamics of individual components, which exert regulatory control in a physiological context, is poorly understood. Now, with our systems-based approach, we reveal kinetic parameters, which define the flow of information through both metabolic and growth-factor arms and thus determine signaling architecture. We also provide a kinetic basis for 1) the advantage of pulsatile-fasted insulin signaling that enables fed-insulin response and 2) the detrimental impact of repeat fed-insulin inputs that causes resistance. Understanding kinetic control of biological processes is as important as identifying components that constitute pathways. Insulin signaling is central for almost all metazoans, and its perturbations are associated with various developmental disorders, metabolic diseases, and aging. While temporal phosphorylation changes and kinetic constants have provided some insights, constant or variable parameters that establish and maintain signal topology are poorly understood. Here, we report kinetic parameters that encode insulin concentration and nutrient-dependent flow of information using iterative experimental and mathematical simulation-based approaches. Our results illustrate how dynamics of distinct phosphorylation events collectively contribute to selective kinetic gating of signals and maximum connectivity of the signaling cascade under normo-insulinemic but not hyper-insulinemic states. In addition to identifying parameters that provide predictive value for maintaining the balance between metabolic and growth-factor arms, we posit a kinetic basis for the emergence of insulin resistance. Given that pulsatile insulin secretion during a fasted state precedes a fed response, our findings reveal rewiring of insulin signaling akin to memory and anticipation, which was hitherto unknown. Striking disparate temporal behavior of key phosphorylation events that destroy the topology under hyper-insulinemic states underscores the importance of unraveling regulatory components that act as bandwidth filters. In conclusion, besides providing fundamental insights, our study will help in identifying therapeutic strategies that conserve coupling between metabolic and growth-factor arms, which is lost in diseases and conditions of hyper-insulinemia.
Collapse
|
24
|
Affiliation(s)
- Karl Z Nadolsky
- Michigan State University College of Human Medicine, Grand Rapids, MI
| | | |
Collapse
|
25
|
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
|
26
|
Laurenti MC, Matveyenko A, Vella A. Measurement of Pulsatile Insulin Secretion: Rationale and Methodology. Metabolites 2021; 11:409. [PMID: 34206296 PMCID: PMC8305896 DOI: 10.3390/metabo11070409] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/29/2022] Open
Abstract
Pancreatic β-cells are responsible for the synthesis and exocytosis of insulin in response to an increase in circulating glucose. Insulin secretion occurs in a pulsatile manner, with oscillatory pulses superimposed on a basal secretion rate. Insulin pulses are a marker of β-cell health, and secretory parameters, such as pulse amplitude, time interval and frequency distribution, are impaired in obesity, aging and type 2 diabetes. In this review, we detail the mechanisms of insulin production and β-cell synchronization that regulate pulsatile insulin secretion, and we discuss the challenges to consider when measuring fast oscillatory secretion in vivo. These include the anatomical difficulties of measuring portal vein insulin noninvasively in humans before the hormone is extracted by the liver and quickly removed from the circulation. Peripheral concentrations of insulin or C-peptide, a peptide cosecreted with insulin, can be used to estimate their secretion profile, but mathematical deconvolution is required. Parametric and nonparametric approaches to the deconvolution problem are evaluated, alongside the assumptions and trade-offs required for their application in the quantification of unknown insulin secretory rates from known peripheral concentrations. Finally, we discuss the therapeutical implication of targeting impaired pulsatile secretion and its diagnostic value as an early indicator of β-cell stress.
Collapse
Affiliation(s)
- Marcello C. Laurenti
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
- Biomedical Engineering and Physiology Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Aleksey Matveyenko
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
| | - Adrian Vella
- Division of Endocrinology, Diabetes & Metabolism, Mayo Clinic, Rochester, MN 55905, USA; (M.C.L.); (A.M.)
| |
Collapse
|
27
|
Saadati M, Jamali Y. The effects of beta-cell mass and function, intercellular coupling, and islet synchrony on [Formula: see text] dynamics. Sci Rep 2021; 11:10268. [PMID: 33986325 PMCID: PMC8119479 DOI: 10.1038/s41598-021-89333-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/26/2021] [Indexed: 11/30/2022] Open
Abstract
Type 2 diabetes (T2D) is a challenging metabolic disorder characterized by a substantial loss of [Formula: see text]-cell mass and alteration of [Formula: see text]-cell function in the islets of Langerhans, disrupting insulin secretion and glucose homeostasis. The mechanisms for deficiency in [Formula: see text]-cell mass and function during the hyperglycemia development and T2D pathogenesis are complex. To study the relative contribution of [Formula: see text]-cell mass to [Formula: see text]-cell function in T2D, we make use of a comprehensive electrophysiological model of human [Formula: see text]-cell clusters. We find that defect in [Formula: see text]-cell mass causes a functional decline in single [Formula: see text]-cell, impairment in intra-islet synchrony, and changes in the form of oscillatory patterns of membrane potential and intracellular [Formula: see text] concentration, which can lead to changes in insulin secretion dynamics and in insulin levels. The model demonstrates a good correspondence between suppression of synchronizing electrical activity and published experimental measurements. We then compare the role of gap junction-mediated electrical coupling with both [Formula: see text]-cell synchronization and metabolic coupling in the behavior of [Formula: see text] concentration dynamics within human islets. Our results indicate that inter-[Formula: see text]-cellular electrical coupling depicts a more important factor in shaping the physiological regulation of islet function and in human T2D. We further predict that varying the whole-cell conductance of delayed rectifier [Formula: see text] channels modifies oscillatory activity patterns of [Formula: see text]-cell population lacking intercellular coupling, which significantly affect [Formula: see text] concentration and insulin secretion.
Collapse
Affiliation(s)
- Maryam Saadati
- Biomathematics Laboratory, Department of Applied Mathematics, School of Mathematical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yousef Jamali
- Biomathematics Laboratory, Department of Applied Mathematics, School of Mathematical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
28
|
Rapid liquid chromatography-mass spectrometry quantitation of glucose-regulating hormones from human islets of Langerhans. J Chromatogr A 2020; 1637:461805. [PMID: 33360778 DOI: 10.1016/j.chroma.2020.461805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Glucose homeostasis is maintained through the secretion of peptide hormones, such as insulin, somatostatin, and glucagon, from islets of Langerhans, clusters of endocrine cells found in the pancreas. This report describes an LC-MS method using multiple reaction monitoring for quantitation of insulin, C-peptide, glucagon, and somatostatin secretion from human islet populations. For rapid analysis, a 5 min separation was achieved using a 2.1 × 30 mm (i.d. x length) C18 column with 2.7 µm diameter core shell particles. A sacrificial protein hydrolysate was used with the sample and found to improve signal magnitude, repeatability, and to reduce carryover between runs. At optimized gradient conditions, the gradient run time was 4.55 min producing an average peak width of 0.3 min, a minimum resolution of 1.2, and a peak capacity of 20. As a proof of concept, the method was used to measure secretions from static incubations of human islets from 2 donors. Insulin and C-peptide were quantified and matched well with literature values of these hormones. We expect that this antibody-free quantitation of multiple hormones secreted from islets will provide insights into the temporal relationships of these peptides in the future.
Collapse
|
29
|
Lee SR, Choi WY, Heo JH, Huh J, Kim G, Lee KP, Kwun HJ, Shin HJ, Baek IJ, Hong EJ. Progesterone increases blood glucose via hepatic progesterone receptor membrane component 1 under limited or impaired action of insulin. Sci Rep 2020; 10:16316. [PMID: 33005004 PMCID: PMC7529793 DOI: 10.1038/s41598-020-73330-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Hepatic gluconeogenesis is the main pathway for blood glucose maintenance activated during fasting. Retardation of insulin action, such as in diabetes mellitus, activates gluconeogenesis during the fed state. While the role of progesterone (P4) in diabetes is controversial, the P4 receptor, progesterone receptor membrane component 1 (PGRMC1), is known to stimulate pancreatic insulin secretion. We investigated the role of P4, via hepatic PGRMC1, during gluconeogenesis. The PGRMC1 binding chemical, AG-205, induced PGRMC1 monomer (25 kDa) abundance, and increased PEPCK expression and glucose production in parallel with cyclic AMP (cAMP) induction in Hep3B cells. PGRMC1-mediated cyclic AMP was inhibited by an adenylate cyclase inhibitor (MDL-12,330A). PEPCK suppression in Pgrmc1 KO hepatocyte was not observed after treatment of MDL-12,330A. PGRMC1 knockdown or overexpression systems in Hep3B cells confirmed that PGRMC1 mediates PEPCK expression via phosphorylation of cAMP-response element binding protein (CREB). CREB phosphorylation and PEPCK expression in primary hepatocytes were greater than that in PGRMC1 knock-out hepatocytes. Progesterone increased PGRMC1 expression, which induced cAMP and PEPCK induction and glucose production. In vivo, P4 suppressed gluconeogenesis following plasma insulin induction under normal conditions in a mouse model. However, P4 increased blood glucose via gluconeogenesis in parallel with increases in PGRMC1 and PEPCK expression in mice in both insulin-deficient and insulin-resistant conditions. We conclude that P4 increases hepatic glucose production via PGRMC1, which may exacerbate hyperglycaemia in diabetes where insulin action is limited.
Collapse
Affiliation(s)
- Sang R Lee
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - Woo-Young Choi
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - Jun H Heo
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - Jiyoung Huh
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - Globinna Kim
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Kyu-Pil Lee
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - Hyo-Jung Kwun
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - Hyun-Jin Shin
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea
| | - In-Jeoung Baek
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Suite 401Veterinary medicine Bldg., Yuseong, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
30
|
Banerjee S, Ghoshal S, Stevens JR, McCommis KS, Gao S, Castro-Sepulveda M, Mizgier ML, Girardet C, Kumar KG, Galgani JE, Niehoff ML, Farr SA, Zhang J, Butler AA. Hepatocyte expression of the micropeptide adropin regulates the liver fasting response and is enhanced by caloric restriction. J Biol Chem 2020; 295:13753-13768. [PMID: 32727846 DOI: 10.1074/jbc.ra120.014381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/22/2020] [Indexed: 12/16/2022] Open
Abstract
The micropeptide adropin encoded by the clock-controlled energy homeostasis-associated gene is implicated in the regulation of glucose metabolism. However, its links to rhythms of nutrient intake, energy balance, and metabolic control remain poorly defined. Using surveys of Gene Expression Omnibus data sets, we confirm that fasting suppresses liver adropin expression in lean C57BL/6J (B6) mice. However, circadian rhythm data are inconsistent. In lean mice, caloric restriction (CR) induces bouts of compulsive binge feeding separated by prolonged fasting intervals, increasing NAD-dependent deacetylase sirtuin-1 signaling important for glucose and lipid metabolism regulation. CR up-regulates adropin expression and induces rhythms correlating with cellular stress-response pathways. Furthermore, adropin expression correlates positively with phosphoenolpyruvate carboxokinase-1 (Pck1) expression, suggesting a link with gluconeogenesis. Our previous data suggest that adropin suppresses gluconeogenesis in hepatocytes. Liver-specific adropin knockout (LAdrKO) mice exhibit increased glucose excursions following pyruvate injections, indicating increased gluconeogenesis. Gluconeogenesis is also increased in primary cultured hepatocytes derived from LAdrKO mice. Analysis of circulating insulin levels and liver expression of fasting-responsive cAMP-dependent protein kinase A (PKA) signaling pathways also suggests enhanced responses in LAdrKO mice during a glucagon tolerance test (250 µg/kg intraperitoneally). Fasting-associated changes in PKA signaling are attenuated in transgenic mice constitutively expressing adropin and in fasting mice treated acutely with adropin peptide. In summary, hepatic adropin expression is regulated by nutrient- and clock-dependent extrahepatic signals. CR induces pronounced postprandial peaks in hepatic adropin expression. Rhythms of hepatic adropin expression appear to link energy balance and cellular stress to the intracellular signal transduction pathways that drive the liver fasting response.
Collapse
Affiliation(s)
- Subhashis Banerjee
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Sarbani Ghoshal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Joseph R Stevens
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Center for Cardiovascular Research, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Saint Louis University Liver Center, Saint Louis University School of Medicine, St. Louis, Missouri USA
| | - Su Gao
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA
| | - Mauricio Castro-Sepulveda
- Laboratorio de Ciencias del Ejercicio. Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| | - Maria L Mizgier
- Departamento de Ciencias de la SaludCarrera de Nutrición y Dietética and Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Clemence Girardet
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - K Ganesh Kumar
- Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA
| | - Jose E Galgani
- Departamento de Ciencias de la SaludCarrera de Nutrición y Dietética and Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michael L Niehoff
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Saint Louis University School of Medicine; Research Service, John Cochran Division, Saint Louis Veterans Affairs Medical Center, Missouri, USA
| | - Susan A Farr
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Division of Geriatric Medicine, Saint Louis University School of Medicine, St. Louis, Saint Louis University School of Medicine; Research Service, John Cochran Division, Saint Louis Veterans Affairs Medical Center, Missouri, USA
| | - Jinsong Zhang
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Andrew A Butler
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA; Department of Metabolism and Aging, Scripps Research Institute, Jupiter, Florida, USA; Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
31
|
Xu SFS, Andersen DB, Izarzugaza JMG, Kuhre RE, Holst JJ. In the rat pancreas, somatostatin tonically inhibits glucagon secretion and is required for glucose-induced inhibition of glucagon secretion. Acta Physiol (Oxf) 2020; 229:e13464. [PMID: 32145704 DOI: 10.1111/apha.13464] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022]
Abstract
AIM It is debated whether the inhibition of glucagon secretion by glucose results from direct effects of glucose on the α-cell (intrinsic regulation) or by paracrine effects exerted by beta- or delta-cell products. METHODS To study this in a more physiological model than isolated islets, we perfused isolated rat pancreases and measured glucagon, insulin and somatostatin secretion in response to graded increases in perfusate glucose concentration (from 3.5 to 4, 5, 6, 7, 8, 10, 12 mmol/L) as well as glucagon responses to blockage/activation of insulin/GABA/somatostatin signalling with or without addition of glucose. RESULTS Glucagon secretion was reduced by about 50% (compared to baseline secretion at 3.5 mmol/L) within minutes after increasing glucose from 4 to 5 mmol/L (P < .01, n = 13). Insulin secretion was increased minimally, but significantly, compared to baseline (3.5 mmol/L) at 4 mmol/L, whereas somatostatin secretion was not significantly increased from baseline until 7 mmol/L. Hereafter secretion of both increased gradually up to 12 mmol/L glucose. Neither recombinant insulin (1 µmol/L), GABA (300 µmol/L) or the insulin-receptor antagonist S961 (at 1 µmol/L) affected basal (3.5 mmol/L) or glucose-induced (5.0 mmol/L) attenuation of glucagon secretion (n = 7-8). Somatostatin-14 attenuated glucagon secretion by ~ 95%, and blockage of somatostatin-receptor (SSTR)-2 or combined blockage of SSTR-2, -3 and -5 by specific antagonists increased glucagon output (at 3.5 mmol/L glucose) and prevented glucose-induced (from 3.5 to 5.0 mmol/L) suppression of secretion. CONCLUSION Somatostatin is a powerful and tonic inhibitor of glucagon secretion from the rat pancreas and is required for glucose to inhibit glucagon secretion.
Collapse
Affiliation(s)
- Stella F. S. Xu
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Daniel B. Andersen
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | | | - Rune E. Kuhre
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Jens J. Holst
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
32
|
Omar-Hmeadi M, Lund PE, Gandasi NR, Tengholm A, Barg S. Paracrine control of α-cell glucagon exocytosis is compromised in human type-2 diabetes. Nat Commun 2020; 11:1896. [PMID: 32312960 PMCID: PMC7171169 DOI: 10.1038/s41467-020-15717-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/23/2020] [Indexed: 01/05/2023] Open
Abstract
Glucagon is released from pancreatic α-cells to activate pathways that raise blood glucose. Its secretion is regulated by α-cell-intrinsic glucose sensing and paracrine control through insulin and somatostatin. To understand the inadequately high glucagon levels that contribute to hyperglycemia in type-2 diabetes (T2D), we analyzed granule behavior, exocytosis and membrane excitability in α-cells of 68 non-diabetic and 21 T2D human donors. We report that exocytosis is moderately reduced in α-cells of T2D donors, without changes in voltage-dependent ion currents or granule trafficking. Dispersed α-cells have a non-physiological V-shaped dose response to glucose, with maximal exocytosis at hyperglycemia. Within intact islets, hyperglycemia instead inhibits α-cell exocytosis, but not in T2D or when paracrine inhibition by insulin or somatostatin is blocked. Surface expression of somatostatin-receptor-2 is reduced in T2D, suggesting a mechanism for the observed somatostatin resistance. Thus, elevated glucagon in human T2D may reflect α-cell insensitivity to paracrine inhibition at hyperglycemia. Glucagon is elevated Type-2 diabetes, which contributes to poor glucose control in patients with the disease. Here the authors report that secretion of the hormone is controlled by paracrine inhibition, and that resistance of α-cells to somatostatin can explain hyperglucagonemia in type-2 diabetes.
Collapse
Affiliation(s)
- Muhmmad Omar-Hmeadi
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Per-Eric Lund
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Nikhil R Gandasi
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Anders Tengholm
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden
| | - Sebastian Barg
- Medical Cell Biology, Uppsala University, Box 571, BMC, 751 23, Uppsala, Sweden.
| |
Collapse
|
33
|
Pozuelo-Sanchez I, Villasanta-Gonzalez A, Alcala-Diaz JF, Vals-Delgado C, Leon-Acuña A, Gonzalez-Requero A, Yubero-Serrano EM, Luque RM, Caballero-Villarraso J, Quesada I, Ordovas JM, Pérez-Martinez P, Roncero-Ramos I, Lopez-Miranda J. Postprandial Lipemia Modulates Pancreatic Alpha-Cell Function in the Prediction of Type 2 Diabetes Development: The CORDIOPREV Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1266-1275. [PMID: 31937103 DOI: 10.1021/acs.jafc.9b06801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Diabetes (T2DM) is a major global health issue, and developing new approaches to its prevention is of paramount importance. We hypothesized that abnormalities in lipid metabolism are involved in alpha-cell deregulation. We therefore studied the metabolic factors underlying alpha-cell dysfunction in T2DM progression after a dietary intervention (Mediterranean and low-fat). Additionally, we evaluated whether postprandial glucagon levels may be considered as a predictive factor of T2DM in cardiovascular patients. Non-T2DM participants from the CORDIOPREV study were categorized by tertiles of the area under the curve (AUC) for triacylglycerols and also by tertiles of AUC for glucagon. Our results showed that patients with higher triacylglycerols levels presented elevated postprandial glucagon (P = 0.009). Moreover, we observed higher risk of T2DM (hazard ratio: 2.65; 95% confidence interval: 1.56-4.53) in subjects with elevated glucagon. In conclusion, high postprandial lipemia may induce alpha-cell dysfunction in cardiovascular patients. Our results also showed that postprandial glucagon levels could be used to predict T2DM development.
Collapse
Affiliation(s)
- Isabel Pozuelo-Sanchez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Alejandro Villasanta-Gonzalez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Juan Francisco Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Cristina Vals-Delgado
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Ana Leon-Acuña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Anabel Gonzalez-Requero
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Elena Maria Yubero-Serrano
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Raul Miguel Luque
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
- Department of Cell Biology, Physiology, and Immunology, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofía University Hospital , University of Córdoba , Córdoba 14004 , Spain
| | | | - Ivan Quesada
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE) , Universidad Miguel Hernández and Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) , Elche 03202 , Spain
| | - José María Ordovas
- Nutrition and Genomics Laboratory , J.M.-US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University , Boston , Massachusetts 02111 , United States
- IMDEA Alimentacion , Madrid 28049 , Spain
| | - Pablo Pérez-Martinez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Irene Roncero-Ramos
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital , University of Cordoba , Córdoba 14004 , Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , Instituto de Salud Carlos III , Madrid 28029 , Spain
| |
Collapse
|
34
|
Roncero-Ramos I, Alcala-Diaz JF, Rangel-Zuñiga OA, Gomez-Delgado F, Jimenez-Lucena R, García-Rios A, Vals-Delgado C, Romero-Baldonado C, Luque RM, Ordovas JM, Perez-Martinez P, Camargo A, Lopez-Miranda J. Prediabetes diagnosis criteria, type 2 diabetes risk and dietary modulation: The CORDIOPREV study. Clin Nutr 2020; 39:492-500. [DOI: 10.1016/j.clnu.2019.02.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 01/21/2019] [Accepted: 02/18/2019] [Indexed: 02/07/2023]
|
35
|
Gilon P. The Role of α-Cells in Islet Function and Glucose Homeostasis in Health and Type 2 Diabetes. J Mol Biol 2020; 432:1367-1394. [PMID: 31954131 DOI: 10.1016/j.jmb.2020.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/23/2019] [Accepted: 01/06/2020] [Indexed: 01/09/2023]
Abstract
Pancreatic α-cells are the major source of glucagon, a hormone that counteracts the hypoglycemic action of insulin and strongly contributes to the correction of acute hypoglycemia. The mechanisms by which glucose controls glucagon secretion are hotly debated, and it is still unclear to what extent this control results from a direct action of glucose on α-cells or is indirectly mediated by β- and/or δ-cells. Besides its hyperglycemic action, glucagon has many other effects, in particular on lipid and amino acid metabolism. Counterintuitively, glucagon seems also required for an optimal insulin secretion in response to glucose by acting on its cognate receptor and, even more importantly, on GLP-1 receptors. Patients with diabetes mellitus display two main alterations of glucagon secretion: a relative hyperglucagonemia that aggravates hyperglycemia, and an impaired glucagon response to hypoglycemia. Under metabolic stress states, such as diabetes, pancreatic α-cells also secrete GLP-1, a glucose-lowering hormone, whereas the gut can produce glucagon. The contribution of extrapancreatic glucagon to the abnormal glucose homeostasis is unclear. Here, I review the possible mechanisms of control of glucagon secretion and the role of α-cells on islet function in healthy state. I discuss the possible causes of the abnormal glucagonemia in diabetes, with particular emphasis on type 2 diabetes, and I briefly comment the current antidiabetic therapies affecting α-cells.
Collapse
Affiliation(s)
- Patrick Gilon
- Université Catholique de Louvain, Institute of Experimental and Clinical Research, Pole of Endocrinology, Diabetes and Nutrition, Avenue Hippocrate 55 (B1.55.06), Brussels, B-1200, Belgium.
| |
Collapse
|
36
|
Human Physiology of Genetic Defects Causing Beta-cell Dysfunction. J Mol Biol 2020; 432:1579-1598. [PMID: 31953147 DOI: 10.1016/j.jmb.2019.12.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
The last decade has revealed hundreds of genetic variants associated with type 2 diabetes, many especially with insulin secretion. However, the evidence for their single or combined effect on beta-cell function relies mostly on genetic association of the variants or genetic risk scores with simple traits, and few have been functionally fully characterized even in cell or animal models. Translating the measured traits into human physiology is not straightforward: none of the various indices for beta-cell function or insulin sensitivity recapitulates the dynamic interplay between glucose sensing, endogenous glucose production, insulin production and secretion, insulin clearance, insulin resistance-to name just a few factors. Because insulin sensitivity is a major determinant of physiological need of insulin, insulin secretion should be evaluated in parallel with insulin sensitivity. On the other hand, multiple physiological or pathogenic processes can either mask or unmask subtle defects in beta-cell function. Even in monogenic diabetes, a clearly pathogenic genetic variant can result in different phenotypic characteristics-or no phenotype at all. In this review, we evaluate the methods available for studying beta-cell function in humans, critically examine the evidence linking some identified variants to a specific beta-cell phenotype, and highlight areas requiring further study.
Collapse
|
37
|
Whitticar NB, Nunemaker CS. Reducing Glucokinase Activity to Enhance Insulin Secretion: A Counterintuitive Theory to Preserve Cellular Function and Glucose Homeostasis. Front Endocrinol (Lausanne) 2020; 11:378. [PMID: 32582035 PMCID: PMC7296051 DOI: 10.3389/fendo.2020.00378] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Pancreatic beta-cells are the only cells in the body that can synthesize and secrete insulin. Through the process of glucose-stimulated insulin secretion, beta-cells release insulin into circulation, stimulating GLUT4-dependent glucose uptake into peripheral tissue. Insulin is normally secreted in pulses that promote signaling at the liver. Long before type 2 diabetes is diagnosed, beta-cells become oversensitive to glucose, causing impaired pulsatility and overstimulation in fasting levels of glucose. The resulting hypersecretion of insulin can cause poor insulin signaling and clearance at the liver, leading to hyperinsulinemia and insulin resistance. Continued overactivity can eventually lead to beta-cell exhaustion and failure at which point type 2 diabetes begins. To prevent or reverse the negative effects of overstimulation, beta-cell activity can be reduced. Clinical studies have revealed the potential of beta-cell rest to reverse new cases of diabetes, but treatments lack durable benefits. In this perspective, we propose an intervention that reduces overactive glucokinase activity in the beta-cell. Glucokinase is known as the glucose sensor of the beta-cell due to its high control over insulin secretion. Therefore, glycolytic overactivity may be responsible for hyperinsulinemia early in the disease and can be reduced to restore normal stimulus-secretion coupling. We have previously reported that reducing glucokinase activity in prediabetic mouse islets can restore pulsatility and enhance insulin secretion. Building on this counterintuitive finding, we review the importance of pulsatile insulin secretion and highlight how normalizing glucose sensing in the beta cell during prediabetic hyperinsulinemia may restore pulsatility and improve glucose homeostasis.
Collapse
Affiliation(s)
- Nicholas B. Whitticar
- Translational Biomedical Sciences Program, Graduate College, Ohio University, Athens, OH, United States
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Craig S. Nunemaker
- Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens OH, United States
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- *Correspondence: Craig S. Nunemaker
| |
Collapse
|
38
|
FCoR-Foxo1 Axis Regulates α-Cell Mass through Repression of Arx Expression. iScience 2019; 23:100798. [PMID: 31923647 PMCID: PMC6951314 DOI: 10.1016/j.isci.2019.100798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/10/2019] [Accepted: 12/19/2019] [Indexed: 01/27/2023] Open
Abstract
Pancreatic endocrine cell development into differentiated α- and β-cells is highly regulated and involves multiple transcription factors. However, the mechanisms behind the determination of α- and β-cell masses remains unclear. We previously identified Foxo1 CoRepressor (FCoR), which inhibits Foxo1 by acetylation. Here we demonstrate that Fcor-knockout mice (FcorKO) exhibit significantly increased α-cell mass, expression of the master α-cell regulatory transcription factor Aristaless-related homeobox (Arx), which can be normalized by β-cell-specific FCoR overexpression (FcorKO-βFcor), and exhibit β-to-α-cell conversion. Compared with FcorKO, β-cell-specific Foxo1 knockout in the FcorKO (DKO) led to decreased Arx expression and α-cell mass. Foxo1 binding to Arx promoter led to DNA methyltransferase 3a (Dnmt3a) dissociation, Arx promoter hypomethylation, and increased Arx expression. In contrast, FCoR suppressed Arx through Foxo1 inhibition and Dnmt3a recruitment to Arx promoter and increased Arx promoter methylation. Our findings suggest that the FCoR-Foxo1 axis regulates pancreatic α-cell mass by suppressing Arx expression.
Collapse
|
39
|
Tengholm A. Cyclic AMP links glucose stimulation to somatostatin secretion in δ-cells. J Gen Physiol 2019; 151:1062-1065. [PMID: 31413066 PMCID: PMC6719405 DOI: 10.1085/jgp.201912417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Tengholm reflects on new work providing insight into the mechanisms of glucose-stimulated somatostatin secretion from δ-cells.
Collapse
Affiliation(s)
- Anders Tengholm
- Department of Medical Cell Biology, Biomedical Centre, Uppsala University
| |
Collapse
|
40
|
Abstract
Controlling the excess and shortage of energy is a fundamental task for living organisms. Diabetes is a representative metabolic disease caused by the malfunction of energy homeostasis. The islets of Langerhans in the pancreas release long-range messengers, hormones, into the blood to regulate the homeostasis of the primary energy fuel, glucose. The hormone and glucose levels in the blood show rhythmic oscillations with a characteristic period of 5-10 min, and the functional roles of the oscillations are not clear. Each islet has [Formula: see text] and [Formula: see text] cells that secrete glucagon and insulin, respectively. These two counter-regulatory hormones appear sufficient to increase and decrease glucose levels. However, pancreatic islets have a third cell type, [Formula: see text] cells, which secrete somatostatin. The three cell populations have a unique spatial organization in islets, and they interact to perturb their hormone secretions. The mini-organs of islets are scattered throughout the exocrine pancreas. Considering that the human pancreas contains approximately a million islets, the coordination of hormone secretion from the multiple sources of islets and cells within the islets should have a significant effect on human physiology. In this review, we introduce the hierarchical organization of tripartite cell networks, and recent biophysical modeling to systematically understand the oscillations and interactions of [Formula: see text], [Formula: see text], and [Formula: see text] cells. Furthermore, we discuss the functional roles and clinical implications of hormonal oscillations and their phase coordination for the diagnosis of type II diabetes.
Collapse
Affiliation(s)
- Taegeun Song
- Department of Physics, Pohang University of Science and Technology, Pohang, Gyeongbuk 37673, Republic of Korea
| | | |
Collapse
|
41
|
Effect of diet composition on insulin sensitivity in humans. Clin Nutr ESPEN 2019; 33:29-38. [PMID: 31451269 DOI: 10.1016/j.clnesp.2019.05.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/21/2019] [Indexed: 12/27/2022]
Abstract
Diet composition has a marked impact on the risk of developing type 2 diabetes and cardiovascular disease. Prospective studies show that dietary patterns with elevated amount of animal products and low quantity of vegetable food items raise the risk of these diseases. In healthy subjects, animal protein intake intensifies insulin resistance whereas plant-based foods enhance insulin sensitivity. Similar effects have been documented in patients with diabetes. Accordingly, pre-pregnancy intake of meat (processed and unprocessed) has been strongly associated with a higher risk of gestational diabetes whereas greater pre-pregnancy vegetable protein consumption is associated with a lower risk of gestational diabetes. Population groups that modify their traditional dietary habit increasing the amount of animal products while reducing plant-based foods experience a remarkable rise in the frequency of type 2 diabetes. The association of animal protein intake with insulin resistance is independent of body mass index. In obese individuals that consume high animal protein diets, insulin sensitivity does not improve following weight loss. Diets aimed to lose weight that encourage restriction of carbohydrates and elevated consumption of animal protein intensify insulin resistance increasing the risk of developing type 2 diabetes and cardiovascular disease. The effect of dietary components on insulin sensitivity may contribute to explain the striking impact of eating habits on the risk of type 2 diabetes and cardiovascular disease. Insulin resistance predisposes to type 2 diabetes in healthy subjects and deteriorates metabolic control in patients with diabetes. In nondiabetic and diabetic individuals, insulin resistance is a major cardiovascular risk factor.
Collapse
|
42
|
Wang C, Xiao Y, Wang J, Hou N, Cui W, Hu X, Zeng F, Yuan Y, Ma D, Sun X, Zhang Y, Zheng W, Liu Y, Shang H, Chen L, Xiao RP, Zhang X. Dynamic changes in insulin and glucagon during disease progression in rhesus monkeys with obesity-related type 2 diabetes mellitus. Diabetes Obes Metab 2019; 21:1111-1120. [PMID: 30575251 DOI: 10.1111/dom.13624] [Citation(s) in RCA: 6] [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: 04/03/2018] [Revised: 12/17/2018] [Accepted: 12/17/2018] [Indexed: 11/28/2022]
Abstract
AIMS To investigate the progression of obesity-related type 2 diabetes mellitus (T2DM) in rhesus monkeys, especially dynamic changes in insulin and glucagon. MATERIALS AND METHODS We followed a cohort of 52 rhesus monkeys for 7 years throughout the progression of obesity-related T2DM. Intravenous glucose tolerance tests were performed every 6 months to evaluate dynamic changes in glucose, insulin and glucagon levels. RESULTS Obesity in rhesus monkeys increased the overall mortality and T2DM morbidity. During the progression of T2DM, glucagon remained consistently elevated, while insulin initially increased in compensation but then dropped to below normal levels when the monkeys developed overt T2DM. After a glucose challenge, both the first and second phases of insulin secretion increased during the early stage of T2DM; in later stages the first phase was delayed and the second phase was diminished. CONCLUSION Our findings showed that, beside the decreased insulin level, hyperglucagonaemia also plays an important role in the development of T2DM.
Collapse
Affiliation(s)
- Can Wang
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yao Xiao
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Jue Wang
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Ning Hou
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Weiyi Cui
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Xiaomin Hu
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Fanxin Zeng
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Ye Yuan
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Dongwei Ma
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Xueting Sun
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yan Zhang
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Wen Zheng
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yuli Liu
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Haibao Shang
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Liangyi Chen
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
43
|
Adeva-Andany MM, Funcasta-Calderón R, Fernández-Fernández C, Castro-Quintela E, Carneiro-Freire N. Metabolic effects of glucagon in humans. J Clin Transl Endocrinol 2019; 15:45-53. [PMID: 30619718 PMCID: PMC6312800 DOI: 10.1016/j.jcte.2018.12.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 01/09/2023] Open
Abstract
Diabetes is a common metabolic disorder that involves glucose, amino acids, and fatty acids. Either insulin deficiency or insulin resistance may cause diabetes. Insulin deficiency causes type 1 diabetes and diabetes associated with total pancreatectomy. Glucagon produces insulin resistance. Glucagon-induced insulin resistance promotes type 2 diabetes and diabetes associated with glucagonoma. Further, glucagon-induced insulin resistance aggravates the metabolic consequences of the insulin-deficient state. A major metabolic effect of insulin is the accumulation of glucose as glycogen in the liver. Glucagon opposes hepatic insulin action and enhances the rate of gluconeogenesis, increasing hepatic glucose output. In order to support gluconeogenesis, glucagon promotes skeletal muscle wasting to supply amino acids as gluconeogenic precursors. Glucagon promotes hepatic fatty acid oxidation to supply energy required to sustain gluconeogenesis. Hepatic fatty acid oxidation generates β-hydroxybutyrate and acetoacetate (ketogenesis). Prospective studies reveal that elevated glucagon secretion at baseline occurs in healthy subjects who develop impaired glucose tolerance at follow-up compared with subjects who maintain normal glucose tolerance, suggesting a relationship between elevated glucagon secretion and development of impaired glucose tolerance. Prospective studies have identified animal protein consumption as an independent risk factor for type 2 diabetes and cardiovascular disease. Animal protein intake activates glucagon secretion inducing sustained elevations in plasma glucagon. Glucagon is a major hormone that causes insulin resistance. Insulin resistance is an established cardiovascular risk factor additionally to its pathogenic role in diabetes. Glucagon may be a potential link between animal protein intake and the risk of developing type 2 diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- María M. Adeva-Andany
- Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| | | | | | | | | |
Collapse
|
44
|
A hindbrain inhibitory microcircuit mediates vagally-coordinated glucose regulation. Sci Rep 2019; 9:2722. [PMID: 30804396 PMCID: PMC6389891 DOI: 10.1038/s41598-019-39490-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
Neurons in the brainstem dorsal vagal complex integrate neural and humoral signals to coordinate autonomic output to viscera that regulate a variety of physiological functions, but how this circuitry regulates metabolism is murky. We tested the hypothesis that premotor, GABAergic neurons in the nucleus tractus solitarius (NTS) form a hindbrain micro-circuit with preganglionic parasympathetic motorneurons of the dorsal motor nucleus of the vagus (DMV) that is capable of modulating systemic blood glucose concentration. In vitro, neuronal activation or inhibition using either excitatory or inhibitory designer receptor exclusively activated by designer drugs (DREADDs) constructs expressed in GABAergic NTS neurons increased or decreased, respectively, action potential firing of GABAergic NTS neurons and downstream synaptic inhibition of the DMV. In vivo, DREADD-mediated activation of GABAergic NTS neurons increased systemic blood glucose concentration, whereas DREADD-mediated silencing of these neurons was without effect. The DREADD-induced hyperglycemia was abolished by blocking peripheral muscarinic receptors, consistent with the hypothesis that altered parasympathetic drive mediated the response. This effect was paralleled by elevated serum glucagon and hepatic phosphoenolpyruvate carboxykinase 1 (PEPCK1) expression, without affecting insulin levels or muscle metabolism. Activity in a hindbrain inhibitory microcircuit is sufficient to modulate systemic glucose concentration, independent of insulin secretion or utilization.
Collapse
|
45
|
Roncero-Ramos I, Jimenez-Lucena R, Alcala-Diaz JF, Vals-Delgado C, Arenas-Larriva AP, Rangel-Zuñiga OA, Leon-Acuña A, Malagon MM, Delgado-Lista J, Perez-Martinez P, Ordovas JM, Camargo A, Lopez-Miranda J. Alpha cell function interacts with diet to modulate prediabetes and Type 2 diabetes. J Nutr Biochem 2018; 62:247-256. [DOI: 10.1016/j.jnutbio.2018.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 08/22/2018] [Accepted: 08/24/2018] [Indexed: 12/19/2022]
|
46
|
Skelin M, Javor E, Lucijanić M, Lucijanić T, Jakupović L, Rahelić D. The role of glucagon in the possible mechanism of cardiovascular mortality reduction in type 2 diabetes patients. Int J Clin Pract 2018; 72:e13274. [PMID: 30295381 DOI: 10.1111/ijcp.13274] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 09/15/2018] [Indexed: 01/10/2023] Open
Abstract
AIM Type 2 diabetes (T2D) is one of the major public health issues worldwide. The main cause of mortality and morbidity among T2D patients are cardiovascular (CV) causes. Various antidiabetics are used in T2D treatment, but until recently they lacked clear evidence of the reduction in CV mortality and all-cause mortality as independent study end-points. The aim of this article was to present and critically evaluate potential mechanisms behind the remarkable results documented in trials with new antidiabetics for the treatment of T2D. METHODS Relevant data were collected using the MEDLINE, PubMed, EMBASE, Web of Science, Science Direct, and Scopus databases with the key words: "type 2 diabetes," "mortality," "glucagon," "empagliflozin," "liraglutide," "insulin" and "QTc." Searches were not limited to specific publication types or study designs. RESULTS The EMPA-REG OUTCOME trial with empagliflozin and LEADER trial with liraglutide presented remarkable results regarding the reduction in mortality in T2D treatment. However, the potential mechanism for those beneficial effects is difficult to determine. It is not likely that improvements in classic CV risk factors are responsible for the observed effect. A potential mechanism may be caused by the elevation of postprandial (PP) glucagon concentrations that can be seen with an empagliflozin and liraglutide therapy, which could have beneficial effects considering the myocardial electrical stability in T2D patients. CONCLUSION This hypothesis throws new light upon possible mechanisms of reduction in mortality in T2D patients.
Collapse
Affiliation(s)
- Marko Skelin
- Department of Pharmacy, General Hospital Šibenik, Šibenik, Croatia
| | - Eugen Javor
- Department of Pharmacy, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia
| | - Marko Lucijanić
- Department of Hematology, Dubrava University Hospital, Zagreb, Croatia
| | - Tomo Lucijanić
- Department of Endocrinology, Diabetes and Clinical Pharmacology, Dubrava University Hospital, Zagreb, Croatia
| | - Lejsa Jakupović
- Healthcare Institution of Community Pharmacy Slavonski Brod, Slavonski Brod, Croatia
| | - Dario Rahelić
- Department of Endocrinology, Diabetes and Clinical Pharmacology, Dubrava University Hospital, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
47
|
Gumbiner B, Esteves B, Dell V, Joh T, Garzone PD, Forgie A, Udata C. Single and multiple ascending-dose study of glucagon-receptor antagonist RN909 in type 2 diabetes: a phase 1, randomized, double-blind, placebo-controlled trial. Endocrine 2018; 62:371-380. [PMID: 30203123 DOI: 10.1007/s12020-018-1597-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE This first-in-human study assessed safety, immunogenicity, pharmacokinetics, and pharmacodynamics of RN909, a monoclonal antibody antagonist of the glucagon receptor, in type 2 diabetes (T2DM) subjects. METHODS This study enrolled 84 T2DM subjects receiving stable metformin regimens. Forty-four subjects were randomized to receive single escalating doses of RN909 (0.3 to 6 mg/kg subcutaneously (SC), or 1 mg/kg intravenously (IV)), or placebo; 40 subjects were randomized to receive multiple escalating doses (50 to 150 mg SC) or placebo every 4 weeks for 12 weeks. RESULTS RN909 was well tolerated; treatment-related elevated liver function tests (LFTs) were observed in 4/33 (12.1%) and 5/32 (15.6%) subjects treated with single and multiple doses, respectively, versus 1/10 (10%) and 0 in the respective placebo groups. RN909 dose-normalized AUCinf increased more than dose-proportionally following single SC doses, and after multiple doses, accumulation ratios ranged from 1.3 to 3.4. The incidence of antidrug antibodies (ADA) was 33% after single doses and 50% after multiple doses. RN909 produced dose-dependent, durable fasting plasma glucose (FPG)-lowering at day 29 (mean change -20.6 to -97.5 mg/dL) and day 85 (mean change; -27.2 to -43.5 mg/dL) after single and multiple doses, respectively. HbA1c also was reduced after single (mean change -0.30% to -1.44%), and multiple doses (-0.83% to -1.56%). CONCLUSION RN909 was well tolerated after single and multiple doses in T2DM subjects, with diarrhea and elevated LFTs the most frequent adverse events. The appearance of ADA did not affect pharmacokinetics or efficacy. Robust lowering of FPG and HbA1c was observed.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Blood Glucose/drug effects
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/immunology
- Dose-Response Relationship, Drug
- Double-Blind Method
- Female
- Humans
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/pharmacokinetics
- Hypoglycemic Agents/therapeutic use
- Male
- Metformin/therapeutic use
- Middle Aged
- Placebos
- Receptors, Glucagon/antagonists & inhibitors
- Receptors, Glucagon/immunology
- Young Adult
Collapse
Affiliation(s)
- Barry Gumbiner
- Pfizer Inc., 10777 Science Center Dr, San Diego, CA, 92121, USA.
| | - Brooke Esteves
- Pfizer Inc., 7 Shipley Circle, Westford, MA, 01886, USA
- ImmunoGen, Inc., 830 Winter Street, Waltham, MA, 02451, USA
| | - Vanessa Dell
- Pfizer Inc. Maine, 235 E. 42nd Street, New York, NY, 10017, USA
| | - Tenshang Joh
- Pfizer Inc., 10646 Science Center Drive, La Jolla, CA, 92121, USA
| | - Pamela D Garzone
- Pfizer Inc., 230 East Grand Ave, South San Francisco, CA, 94080, USA
| | - Alison Forgie
- Pfizer Inc., 10646 Science Center Drive, La Jolla, CA, 92121, USA
| | | |
Collapse
|
48
|
Petrenko V, Philippe J, Dibner C. Time zones of pancreatic islet metabolism. Diabetes Obes Metab 2018; 20 Suppl 2:116-126. [PMID: 30230177 DOI: 10.1111/dom.13383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/04/2018] [Accepted: 05/23/2018] [Indexed: 12/28/2022]
Abstract
Most living beings possess an intrinsic system of circadian oscillators, allowing anticipation of the Earth's rotation around its own axis. The mammalian circadian timing system orchestrates nearly all aspects of physiology and behaviour. Together with systemic signals originating from the central clock that resides in the hypothalamic suprachiasmatic nucleus, peripheral oscillators orchestrate tissue-specific fluctuations in gene transcription and translation, and posttranslational modifications, driving overt rhythms in physiology and behaviour. There is accumulating evidence of a reciprocal connection between the circadian oscillator and most aspects of physiology and metabolism, in particular as the circadian system plays a critical role in orchestrating body glucose homeostasis. Recent reports imply that circadian clocks operative in the endocrine pancreas regulate insulin secretion, and that islet clock perturbation in rodents leads to the development of overt type 2 diabetes. While whole islet clocks have been extensively studied during the last years, the heterogeneity of islet cell oscillators and the interplay between α- and β-cellular clocks for orchestrating glucagon and insulin secretion have only recently gained attention. Here, we review recent findings on the molecular makeup of the circadian clocks operative in pancreatic islet cells in rodents and in humans, and focus on the physiologically relevant synchronizers that are resetting these time-keepers. Moreover, the implication of islet clock functional outputs in the temporal coordination of metabolism in health and disease will be highlighted.
Collapse
Affiliation(s)
- Volodymyr Petrenko
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| | - Charna Dibner
- Division of Endocrinology, Diabetes, Hypertension and Nutrition, Department of Internal Medicine Specialties, University Hospital of Geneva, Geneva, Switzerland
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Institute of Genetics and Genomics in Geneva (iGE3), Geneva, Switzerland
| |
Collapse
|
49
|
MafB Is Critical for Glucagon Production and Secretion in Mouse Pancreatic α Cells In Vivo. Mol Cell Biol 2018; 38:MCB.00504-17. [PMID: 29378833 DOI: 10.1128/mcb.00504-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
The MafB transcription factor is expressed in pancreatic α and β cells during development but becomes exclusive to α cells in adult rodents. Mafb-null (Mafb-/- ) mice were reported to have reduced α- and β-cell numbers throughout embryonic development. To further analyze the postnatal function of MafB in the pancreas, we generated endocrine cell-specific (MafbΔEndo ) and tamoxifen-dependent (MafbΔTAM ) Mafb knockout mice. MafbΔEndo mice exhibited reduced populations of insulin-positive (insulin+) and glucagon+ cells at postnatal day 0, but the insulin+ cell population recovered by 8 weeks of age. In contrast, the Arx+ glucagon+ cell fraction and glucagon expression remained decreased even in adulthood. MafbΔTAM mice, with Mafb deleted after pancreas maturation, also demonstrated diminished glucagon+ cells and glucagon content without affecting β cells. A decreased Arx+ glucagon+ cell population in MafbΔEndo mice was compensated for by an increased Arx+ pancreatic polypeptide+ cell population. Furthermore, gene expression analyses from both MafbΔEndo and MafbΔTAM islets revealed that MafB is a key regulator of glucagon expression in α cells. Finally, both mutants failed to respond to arginine, likely due to impaired arginine transporter gene expression and glucagon production ability. Taken together, our findings reveal that MafB is critical for the functional maintenance of mouse α cells in vivo, including glucagon production and secretion, as well as in development.
Collapse
|
50
|
Lei CL, Kellard JA, Hara M, Johnson JD, Rodriguez B, Briant LJ. Beta-cell hubs maintain Ca 2+ oscillations in human and mouse islet simulations. Islets 2018; 10:151-167. [PMID: 30142036 PMCID: PMC6113907 DOI: 10.1080/19382014.2018.1493316] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 06/22/2018] [Indexed: 12/17/2022] Open
Abstract
Islet β-cells are responsible for secreting all circulating insulin in response to rising plasma glucose concentrations. These cells are a phenotypically diverse population that express great functional heterogeneity. In mice, certain β-cells (termed 'hubs') have been shown to be crucial for dictating the islet response to high glucose, with inhibition of these hub cells abolishing the coordinated Ca2+ oscillations necessary for driving insulin secretion. These β-cell hubs were found to be highly metabolic and susceptible to pro-inflammatory and glucolipotoxic insults. In this study, we explored the importance of hub cells in human by constructing mathematical models of Ca2+ activity in human islets. Our simulations revealed that hubs dictate the coordinated Ca2+ response in both mouse and human islets; silencing a small proportion of hubs abolished whole-islet Ca2+ activity. We also observed that if hubs are assumed to be preferentially gap junction coupled, then the simulations better adhere to the available experimental data. Our simulations of 16 size-matched mouse and human islet architectures revealed that there are species differences in the role of hubs; Ca2+ activity in human islets was more vulnerable to hub inhibition than mouse islets. These simulation results not only substantiate the existence of β-cell hubs, but also suggest that hubs may be favorably coupled in the electrical and metabolic network of the islet, and that targeted destruction of these cells would greatly impair human islet function.
Collapse
Affiliation(s)
- Chon-Lok Lei
- Doctoral Training Centre, University of Oxford, Oxford, UK
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Joely A. Kellard
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, USA
| | - James D. Johnson
- Department of Cellular and Physiological Sciences, Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, UK
| | - Linford J.B. Briant
- Department of Computer Science, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| |
Collapse
|