1
|
Šterk M, Zhang Y, Pohorec V, Leitgeb EP, Dolenšek J, Benninger RKP, Stožer A, Kravets V, Gosak M. Network representation of multicellular activity in pancreatic islets: Technical considerations for functional connectivity analysis. PLoS Comput Biol 2024; 20:e1012130. [PMID: 38739680 PMCID: PMC11115366 DOI: 10.1371/journal.pcbi.1012130] [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: 01/03/2024] [Revised: 05/23/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
Within the islets of Langerhans, beta cells orchestrate synchronized insulin secretion, a pivotal aspect of metabolic homeostasis. Despite the inherent heterogeneity and multimodal activity of individual cells, intercellular coupling acts as a homogenizing force, enabling coordinated responses through the propagation of intercellular waves. Disruptions in this coordination are implicated in irregular insulin secretion, a hallmark of diabetes. Recently, innovative approaches, such as integrating multicellular calcium imaging with network analysis, have emerged for a quantitative assessment of the cellular activity in islets. However, different groups use distinct experimental preparations, microscopic techniques, apply different methods to process the measured signals and use various methods to derive functional connectivity patterns. This makes comparisons between findings and their integration into a bigger picture difficult and has led to disputes in functional connectivity interpretations. To address these issues, we present here a systematic analysis of how different approaches influence the network representation of islet activity. Our findings show that the choice of methods used to construct networks is not crucial, although care is needed when combining data from different islets. Conversely, the conclusions drawn from network analysis can be heavily affected by the pre-processing of the time series, the type of the oscillatory component in the signals, and by the experimental preparation. Our tutorial-like investigation aims to resolve interpretational issues, reconcile conflicting views, advance functional implications, and encourage researchers to adopt connectivity analysis. As we conclude, we outline challenges for future research, emphasizing the broader applicability of our conclusions to other tissues exhibiting complex multicellular dynamics.
Collapse
Affiliation(s)
- Marko Šterk
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Yaowen Zhang
- Department of Pediatrics, Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
| | - Viljem Pohorec
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | | - Jurij Dolenšek
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Richard K. P. Benninger
- Department of Bioengineering, Barbara Davis Center for Diabetes, Aurora, Colorado, United States of America
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Vira Kravets
- Department of Pediatrics, Department of Bioengineering, University of California, San Diego, La Jolla, California, United States of America
- Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, California, United States of America
| | - Marko Gosak
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea, Maribor
| |
Collapse
|
2
|
Lu Y, Xu J, Li Y, Wang R, Dai C, Zhang B, Zhang X, Xu L, Tao Y, Han M, Guo R, Wu Q, Wu L, Meng Z, Tan M, Li J. DRAK2 suppresses autophagy by phosphorylating ULK1 at Ser 56 to diminish pancreatic β cell function upon overnutrition. Sci Transl Med 2024; 16:eade8647. [PMID: 38324636 DOI: 10.1126/scitranslmed.ade8647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/12/2024] [Indexed: 02/09/2024]
Abstract
Impeded autophagy can impair pancreatic β cell function by causing apoptosis, of which DAP-related apoptosis-inducing kinase-2 (DRAK2) is a critical regulator. Here, we identified a marked up-regulation of DRAK2 in pancreatic tissue across humans, macaques, and mice with type 2 diabetes (T2D). Further studies in mice showed that conditional knockout (cKO) of DRAK2 in pancreatic β cells protected β cell function against high-fat diet feeding along with sustained autophagy and mitochondrial function. Phosphoproteome analysis in isolated mouse primary islets revealed that DRAK2 directly phosphorylated unc-51-like autophagy activating kinase 1 (ULK1) at Ser56, which was subsequently found to induce ULK1 ubiquitylation and suppress autophagy. ULK1-S56A mutation or pharmacological inhibition of DRAK2 preserved mitochondrial function and insulin secretion against lipotoxicity in mouse primary islets, Min6 cells, or INS-1E cells. In conclusion, these findings together indicate an indispensable role of the DRAK2-ULK1 axis in pancreatic β cells upon metabolic challenge, which offers a potential target to protect β cell function in T2D.
Collapse
Affiliation(s)
- Yuting Lu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Junyu Xu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P. R. China
| | - Yufeng Li
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Ruoran Wang
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
| | - Chengqiu Dai
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bingqian Zhang
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xinwen Zhang
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Lei Xu
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P. R. China
| | - Yunhua Tao
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Ming Han
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Ren Guo
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
| | - Qingqian Wu
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
| | - Linshi Wu
- Shanghai Jiaotong University, School of Medicine, Renji Hospital, Shanghai, 201112, P. R. China
| | - Zhuoxian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, P. R. China
| | - Minjia Tan
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jingya Li
- State Key Laboratory of Drug Research, the National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences. Shanghai, 201203, P. R. China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
3
|
Paradiž Leitgeb E, Kerčmar J, Križančić Bombek L, Pohorec V, Skelin Klemen M, Slak Rupnik M, Gosak M, Dolenšek J, Stožer A. Exendin-4 affects calcium signalling predominantly during activation and activity of beta cell networks in acute mouse pancreas tissue slices. Front Endocrinol (Lausanne) 2024; 14:1315520. [PMID: 38292770 PMCID: PMC10826511 DOI: 10.3389/fendo.2023.1315520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
Tight control of beta cell stimulus-secretion coupling is crucial for maintaining homeostasis of energy-rich nutrients. While glucose serves as a primary regulator of this process, incretins augment beta cell function, partly by enhancing cytosolic [Ca2+] dynamics. However, the details of how precisely they affect beta cell recruitment during activation, their active time, and functional connectivity during plateau activity, and how they influence beta cell deactivation remain to be described. Performing functional multicellular Ca2+ imaging in acute mouse pancreas tissue slices enabled us to systematically assess the effects of the GLP-1 receptor agonist exendin-4 (Ex-4) simultaneously in many coupled beta cells with high resolution. In otherwise substimulatory glucose, Ex-4 was able to recruit approximately a quarter of beta cells into an active state. Costimulation with Ex-4 and stimulatory glucose shortened the activation delays and accelerated beta cell activation dynamics. More specifically, active time increased faster, and the time required to reach half-maximal activation was effectively halved in the presence of Ex-4. Moreover, the active time and regularity of [Ca2+]IC oscillations increased, especially during the first part of beta cell response. In contrast, subsequent addition of Ex-4 to already active cells did not significantly enhance beta cell activity. Network analyses further confirmed increased connectivity during activation and activity in the presence of Ex-4, with hub cell roles remaining rather stable in both control experiments and experiments with Ex-4. Interestingly, Ex-4 demonstrated a biphasic effect on deactivation, slightly prolonging beta cell activity at physiological concentrations and shortening deactivation delays at supraphysiological concentrations. In sum, costimulation by Ex-4 and glucose increases [Ca2+]IC during beta cell activation and activity, indicating that the effect of incretins may, to an important extent, be explained by enhanced [Ca2+]IC signals. During deactivation, previous incretin stimulation does not critically prolong cellular activity, which corroborates their low risk of hypoglycemia.
Collapse
Affiliation(s)
- Eva Paradiž Leitgeb
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jasmina Kerčmar
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | | | - Vilijem Pohorec
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Alma Mater Europaea-European Center Maribor, Maribor, Slovenia
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Alma Mater Europaea-European Center Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
4
|
Šterk M, Barać U, Stožer A, Gosak M. Both electrical and metabolic coupling shape the collective multimodal activity and functional connectivity patterns in beta cell collectives: A computational model perspective. Phys Rev E 2023; 108:054409. [PMID: 38115462 DOI: 10.1103/physreve.108.054409] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 10/20/2023] [Indexed: 12/21/2023]
Abstract
Pancreatic beta cells are coupled excitable oscillators that synchronize their activity via different communication pathways. Their oscillatory activity manifests itself on multiple timescales and consists of bursting electrical activity, subsequent oscillations in the intracellular Ca^{2+}, as well as oscillations in metabolism and exocytosis. The coordination of the intricate activity on the multicellular level plays a key role in the regulation of physiological pulsatile insulin secretion and is incompletely understood. In this paper, we investigate theoretically the principles that give rise to the synchronized activity of beta cell populations by building up a phenomenological multicellular model that incorporates the basic features of beta cell dynamics. Specifically, the model is composed of coupled slow and fast oscillatory units that reflect metabolic processes and electrical activity, respectively. Using a realistic description of the intercellular interactions, we study how the combination of electrical and metabolic coupling generates collective rhythmicity and shapes functional beta cell networks. It turns out that while electrical coupling solely can synchronize the responses, the addition of metabolic interactions further enhances coordination, the spatial range of interactions increases the number of connections in the functional beta cell networks, and ensures a better consistency with experimental findings. Moreover, our computational results provide additional insights into the relationship between beta cell heterogeneity, their activity profiles, and functional connectivity, supplementing thereby recent experimental results on endocrine networks.
Collapse
Affiliation(s)
- Marko Šterk
- Department of Physics, Faculty of Natural Sciences and Mathematics, Koroška cesta 160, University of Maribor, 2000 Maribor, Slovenia
- Institute of Physiology, Faculty of Medicine, Taborska ulica 8, University of Maribor, 2000 Maribor, Slovenia
- Alma Mater Europaea, Slovenska ulica 17, 2000 Maribor, Slovenia
| | - Uroš Barać
- Department of Physics, Faculty of Natural Sciences and Mathematics, Koroška cesta 160, University of Maribor, 2000 Maribor, Slovenia
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, Taborska ulica 8, University of Maribor, 2000 Maribor, Slovenia
| | - Marko Gosak
- Department of Physics, Faculty of Natural Sciences and Mathematics, Koroška cesta 160, University of Maribor, 2000 Maribor, Slovenia
- Institute of Physiology, Faculty of Medicine, Taborska ulica 8, University of Maribor, 2000 Maribor, Slovenia
- Alma Mater Europaea, Slovenska ulica 17, 2000 Maribor, Slovenia
| |
Collapse
|
5
|
Freyberg Z, Gittes GK. Roles of Pancreatic Islet Catecholamine Neurotransmitters in Glycemic Control and in Antipsychotic Drug-Induced Dysglycemia. Diabetes 2023; 72:3-15. [PMID: 36538602 PMCID: PMC9797319 DOI: 10.2337/db22-0522] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/24/2022] [Indexed: 12/24/2022]
Abstract
Catecholamine neurotransmitters dopamine (DA) and norepinephrine (NE) are essential for a myriad of functions throughout the central nervous system, including metabolic regulation. These molecules are also present in the pancreas, and their study may shed light on the effects of peripheral neurotransmission on glycemic control. Though sympathetic innervation to islets provides NE that signals at local α-cell and β-cell adrenergic receptors to modify hormone secretion, α-cells and β-cells also synthesize catecholamines locally. We propose a model where α-cells and β-cells take up catecholamine precursors in response to postprandial availability, preferentially synthesizing DA. The newly synthesized DA signals in an autocrine/paracrine manner to regulate insulin and glucagon secretion and maintain glycemic control. This enables islets to couple local catecholamine signaling to changes in nutritional state. We also contend that the DA receptors expressed by α-cells and β-cells are targeted by antipsychotic drugs (APDs)-some of the most widely prescribed medications today. Blockade of local DA signaling contributes significantly to APD-induced dysglycemia, a major contributor to treatment discontinuation and development of diabetes. Thus, elucidating the peripheral actions of catecholamines will provide new insights into the regulation of metabolic pathways and may lead to novel, more effective strategies to tune metabolism and treat diabetes.
Collapse
Affiliation(s)
- Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA
| | - George K. Gittes
- Division of Pediatric Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
6
|
Gosak M, Yan-Do R, Lin H, MacDonald PE, Stožer A. Ca2+ Oscillations, Waves, and Networks in Islets From Human Donors With and Without Type 2 Diabetes. Diabetes 2022; 71:2584-2596. [PMID: 36084321 PMCID: PMC9750953 DOI: 10.2337/db22-0004] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/01/2022] [Indexed: 01/11/2023]
Abstract
Pancreatic islets are highly interconnected structures that produce pulses of insulin and other hormones, maintaining normal homeostasis of glucose and other nutrients. Normal stimulus-secretion and intercellular coupling are essential to regulated secretory responses, and these hallmarks are known to be altered in diabetes. In the current study, we used calcium imaging of isolated human islets to assess their collective behavior. The activity occurred in the form of calcium oscillations, was synchronized across different regions of islets through calcium waves, and was glucose dependent: higher glucose enhanced the activity, elicited a greater proportion of global calcium waves, and led to denser and less fragmented functional networks. Hub regions were identified in stimulatory conditions, and they were characterized by long active times. Moreover, calcium waves were found to be initiated in different subregions and the roles of initiators and hubs did not overlap. In type 2 diabetes, glucose dependence was retained, but reduced activity, locally restricted waves, and more segregated networks were detected compared with control islets. Interestingly, hub regions seemed to suffer the most by losing a disproportionately large fraction of connections. These changes affected islets from donors with diabetes in a heterogeneous manner.
Collapse
Affiliation(s)
- Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Richard Yan-Do
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Haopeng Lin
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Patrick E. MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
7
|
Wnt4 is heterogeneously activated in maturing β-cells to control calcium signaling, metabolism and function. Nat Commun 2022; 13:6255. [PMID: 36271049 PMCID: PMC9587236 DOI: 10.1038/s41467-022-33841-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/05/2022] [Indexed: 12/25/2022] Open
Abstract
Diabetes is a multifactorial disorder characterized by loss or dysfunction of pancreatic β-cells. β-cells are heterogeneous, exhibiting different glucose sensing, insulin secretion and gene expression. They communicate with other endocrine cell types via paracrine signals and between β-cells via gap junctions. Here, we identify the importance of signaling between β-cells via the extracellular signal WNT4. We show heterogeneity in Wnt4 expression, most strikingly in the postnatal maturation period, Wnt4-positive cells, being more mature while Wnt4-negative cells are more proliferative. Knock-out in adult β-cells shows that WNT4 controls the activation of calcium signaling in response to a glucose challenge, as well as metabolic pathways converging to lower ATP/ADP ratios, thereby reducing insulin secretion. These results reveal that paracrine signaling between β-cells is important in addition to gap junctions in controling insulin secretion. Together with previous reports of WNT4 up-regulation in obesity our observations suggest an adaptive insulin response coordinating β-cells.
Collapse
|
8
|
Stožer A, Šterk M, Paradiž Leitgeb E, Markovič R, Skelin Klemen M, Ellis CE, Križančić Bombek L, Dolenšek J, MacDonald PE, Gosak M. From Isles of Königsberg to Islets of Langerhans: Examining the Function of the Endocrine Pancreas Through Network Science. Front Endocrinol (Lausanne) 2022; 13:922640. [PMID: 35784543 PMCID: PMC9240343 DOI: 10.3389/fendo.2022.922640] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Islets of Langerhans are multicellular microorgans located in the pancreas that play a central role in whole-body energy homeostasis. Through secretion of insulin and other hormones they regulate postprandial storage and interprandial usage of energy-rich nutrients. In these clusters of hormone-secreting endocrine cells, intricate cell-cell communication is essential for proper function. Electrical coupling between the insulin-secreting beta cells through gap junctions composed of connexin36 is particularly important, as it provides the required, most important, basis for coordinated responses of the beta cell population. The increasing evidence that gap-junctional communication and its modulation are vital to well-regulated secretion of insulin has stimulated immense interest in how subpopulations of heterogeneous beta cells are functionally arranged throughout the islets and how they mediate intercellular signals. In the last decade, several novel techniques have been proposed to assess cooperation between cells in islets, including the prosperous combination of multicellular imaging and network science. In the present contribution, we review recent advances related to the application of complex network approaches to uncover the functional connectivity patterns among cells within the islets. We first provide an accessible introduction to the basic principles of network theory, enumerating the measures characterizing the intercellular interactions and quantifying the functional integration and segregation of a multicellular system. Then we describe methodological approaches to construct functional beta cell networks, point out possible pitfalls, and specify the functional implications of beta cell network examinations. We continue by highlighting the recent findings obtained through advanced multicellular imaging techniques supported by network-based analyses, giving special emphasis to the current developments in both mouse and human islets, as well as outlining challenges offered by the multilayer network formalism in exploring the collective activity of islet cell populations. Finally, we emphasize that the combination of these imaging techniques and network-based analyses does not only represent an innovative concept that can be used to describe and interpret the physiology of islets, but also provides fertile ground for delineating normal from pathological function and for quantifying the changes in islet communication networks associated with the development of diabetes mellitus.
Collapse
Affiliation(s)
- Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marko Šterk
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Eva Paradiž Leitgeb
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rene Markovič
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Institute of Mathematics and Physics, Faculty of Electrical Engineering and Computer Science, University of Maribor, Maribor, Slovenia
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Cara E. Ellis
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Patrick E. MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
9
|
Collares-Buzato CB, Carvalho CP. Is type 2 diabetes mellitus another intercellular junction-related disorder? Exp Biol Med (Maywood) 2022; 247:743-755. [PMID: 35466731 DOI: 10.1177/15353702221090464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2D) is nowadays a worldwide epidemic and has become a major challenge for health systems around the world. It is a multifactorial disorder, characterized by a chronic state of hyperglycemia caused by defects in the production as well as in the peripheral action of insulin. This minireview highlights the experimental and clinical evidence that supports the novel idea that intercellular junctions (IJs)-mediated cell-cell contacts play a role in the pathogenesis of T2D. It focuses on IJs repercussion for endocrine pancreas, intestinal barrier, and kidney dysfunctions that contribute to the onset and evolution of this metabolic disorder.
Collapse
Affiliation(s)
- Carla B Collares-Buzato
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, CEP 13083-970, Brazil
| | - Carolina Pf Carvalho
- Department of Biosciences, Federal University of São Paulo (UNIFESP), Santos, SP, CEP 11015-020, Brazil
| |
Collapse
|
10
|
Guérineau NC, Campos P, Le Tissier PR, Hodson DJ, Mollard P. Cell Networks in Endocrine/Neuroendocrine Gland Function. Compr Physiol 2022; 12:3371-3415. [PMID: 35578964 DOI: 10.1002/cphy.c210031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reproduction, growth, stress, and metabolism are determined by endocrine/neuroendocrine systems that regulate circulating hormone concentrations. All these systems generate rhythms and changes in hormone pulsatility observed in a variety of pathophysiological states. Thus, the output of endocrine/neuroendocrine systems must be regulated within a narrow window of effective hormone concentrations but must also maintain a capacity for plasticity to respond to changing physiological demands. Remarkably most endocrinologists still have a "textbook" view of endocrine gland organization which has emanated from 20th century histological studies on thin 2D tissue sections. However, 21st -century technological advances, including in-depth 3D imaging of specific cell types have vastly changed our knowledge. We now know that various levels of multicellular organization can be found across different glands, that organizational motifs can vary between species and can be modified to enhance or decrease hormonal release. This article focuses on how the organization of cells regulates hormone output using three endocrine/neuroendocrine glands that present different levels of organization and complexity: the adrenal medulla, with a single neuroendocrine cell type; the anterior pituitary, with multiple intermingled cell types; and the pancreas with multiple intermingled cell types organized into distinct functional units. We give an overview of recent methodologies that allow the study of the different components within endocrine systems, particularly their temporal and spatial relationships. We believe the emerging findings about network organization, and its impact on hormone secretion, are crucial to understanding how homeostatic regulation of endocrine axes is carried out within endocrine organs themselves. © 2022 American Physiological Society. Compr Physiol 12:3371-3415, 2022.
Collapse
Affiliation(s)
| | - Pauline Campos
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Paul R Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,COMPARE University of Birmingham and University of Nottingham Midlands, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patrice Mollard
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
11
|
Akalestou E, Suba K, Lopez-Noriega L, Georgiadou E, Chabosseau P, Gallie A, Wretlind A, Legido-Quigley C, Leclerc I, Salem V, Rutter GA. Intravital imaging of islet Ca 2+ dynamics reveals enhanced β cell connectivity after bariatric surgery in mice. Nat Commun 2021; 12:5165. [PMID: 34453049 PMCID: PMC8397709 DOI: 10.1038/s41467-021-25423-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/06/2021] [Indexed: 11/25/2022] Open
Abstract
Bariatric surgery improves both insulin sensitivity and secretion and can induce diabetes remission. However, the mechanisms and time courses of these changes, particularly the impact on β cell function, are difficult to monitor directly. In this study, we investigated the effect of Vertical Sleeve Gastrectomy (VSG) on β cell function in vivo by imaging Ca2+ dynamics in islets engrafted into the anterior eye chamber. Mirroring its clinical utility, VSG in mice results in significantly improved glucose tolerance, and enhanced insulin secretion. We reveal that these benefits are underpinned by augmented β cell function and coordinated activity across the islet. These effects involve changes in circulating GLP-1 levels which may act both directly and indirectly on the β cell, in the latter case through changes in body weight. Thus, bariatric surgery leads to time-dependent increases in β cell function and intra-islet connectivity which are likely to contribute to diabetes remission.
Collapse
Affiliation(s)
- Elina Akalestou
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Kinga Suba
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Livia Lopez-Noriega
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Eleni Georgiadou
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Pauline Chabosseau
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Alasdair Gallie
- grid.413629.b0000 0001 0705 4923Central Biological Services (CBS) Hammersmith Hospital Campus, London, UK
| | - Asger Wretlind
- grid.419658.70000 0004 0646 7285Systems Medicine, Steno Diabetes Center, Gentofte, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- grid.419658.70000 0004 0646 7285Systems Medicine, Steno Diabetes Center, Gentofte, Copenhagen, Denmark
| | - Isabelle Leclerc
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Victoria Salem
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK ,grid.413629.b0000 0001 0705 4923Section of Investigative Medicine, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Guy A. Rutter
- grid.413629.b0000 0001 0705 4923Section of Cell Biology and Functional Genomics, Imperial College London, Hammersmith Hospital Campus, London, UK ,grid.59025.3b0000 0001 2224 0361Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore, Singapore ,grid.14848.310000 0001 2292 3357Centre de Recherches du CHUM, University of Montreal, Montreal, QC Canada
| |
Collapse
|
12
|
Stožer A, Skelin Klemen M, Gosak M, Križančić Bombek L, Pohorec V, Slak Rupnik M, Dolenšek J. Glucose-dependent activation, activity, and deactivation of beta cell networks in acute mouse pancreas tissue slices. Am J Physiol Endocrinol Metab 2021; 321:E305-E323. [PMID: 34280052 DOI: 10.1152/ajpendo.00043.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022]
Abstract
Many details of glucose-stimulated intracellular calcium changes in β cells during activation, activity, and deactivation, as well as their concentration-dependence, remain to be analyzed. Classical physiological experiments indicated that in islets, functional differences between individual cells are largely attenuated, but recent findings suggest considerable intercellular heterogeneity, with some cells possibly coordinating the collective responses. To address the above with an emphasis on heterogeneity and describing the relations between classical physiological and functional network properties, we performed functional multicellular calcium imaging in mouse pancreas tissue slices over a wide range of glucose concentrations. During activation, delays to activation of cells and any-cell-to-first-responder delays are shortened, and the sizes of simultaneously responding clusters increased with increasing glucose concentrations. Exactly the opposite characterized deactivation. The frequency of fast calcium oscillations during activity increased with increasing glucose up to 12 mM glucose concentration, beyond which oscillation duration became longer, resulting in a homogenous increase in active time. In terms of functional connectivity, islets progressed from a very segregated network to a single large functional unit with increasing glucose concentration. A comparison between classical physiological and network parameters revealed that the first-responders during activation had longer active times during plateau and the most active cells during the plateau tended to deactivate later. Cells with the most functional connections tended to activate sooner, have longer active times, and deactivate later. Our findings provide a common ground for recent differing views on β cell heterogeneity and an important baseline for future studies of stimulus-secretion and intercellular coupling.NEW & NOTEWORTHY We assessed concentration-dependence in coupled β cells, degree of functional heterogeneity, and uncovered possible specialized subpopulations during the different phases of the response to glucose at the level of many individual cells. To this aim, we combined acute mouse pancreas tissue slices with functional multicellular calcium imaging over a wide range from threshold (7 mM) and physiological (8 and 9 mM) to supraphysiological (12 and 16 mM) glucose concentrations, classical physiological, and advanced network analyses.
Collapse
Affiliation(s)
- Andraž Stožer
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Maša Skelin Klemen
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Marko Gosak
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | | | - Viljem Pohorec
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Alma Mater Europaea-European Center Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| |
Collapse
|
13
|
Stožer A, Paradiž Leitgeb E, Pohorec V, Dolenšek J, Križančić Bombek L, Gosak M, Skelin Klemen M. The Role of cAMP in Beta Cell Stimulus-Secretion and Intercellular Coupling. Cells 2021; 10:1658. [PMID: 34359828 PMCID: PMC8304079 DOI: 10.3390/cells10071658] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 12/22/2022] Open
Abstract
Pancreatic beta cells secrete insulin in response to stimulation with glucose and other nutrients, and impaired insulin secretion plays a central role in development of diabetes mellitus. Pharmacological management of diabetes includes various antidiabetic drugs, including incretins. The incretin hormones, glucagon-like peptide-1 and gastric inhibitory polypeptide, potentiate glucose-stimulated insulin secretion by binding to G protein-coupled receptors, resulting in stimulation of adenylate cyclase and production of the secondary messenger cAMP, which exerts its intracellular effects through activation of protein kinase A or the guanine nucleotide exchange protein 2A. The molecular mechanisms behind these two downstream signaling arms are still not fully elucidated and involve many steps in the stimulus-secretion coupling cascade, ranging from the proximal regulation of ion channel activity to the central Ca2+ signal and the most distal exocytosis. In addition to modifying intracellular coupling, the effect of cAMP on insulin secretion could also be at least partly explained by the impact on intercellular coupling. In this review, we systematically describe the possible roles of cAMP at these intra- and inter-cellular signaling nodes, keeping in mind the relevance for the whole organism and translation to humans.
Collapse
Affiliation(s)
- Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Eva Paradiž Leitgeb
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Viljem Pohorec
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
- Faculty of Natural Sciences and Mathematics, University of Maribor, SI-2000 Maribor, Slovenia
| | - Lidija Križančić Bombek
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
- Faculty of Natural Sciences and Mathematics, University of Maribor, SI-2000 Maribor, Slovenia
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; (A.S.); (E.P.L.); (V.P.); (J.D.); (L.K.B.); (M.G.)
| |
Collapse
|
14
|
de Jesus DS, Mak TCS, Wang YF, von Ohlen Y, Bai Y, Kane E, Chabosseau P, Chahrour CM, Distaso W, Salem V, Tomas A, Stoffel M, Rutter GA, Latreille M. Dysregulation of the Pdx1/Ovol2/Zeb2 axis in dedifferentiated β-cells triggers the induction of genes associated with epithelial-mesenchymal transition in diabetes. Mol Metab 2021; 53:101248. [PMID: 33989778 PMCID: PMC8184664 DOI: 10.1016/j.molmet.2021.101248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/24/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE β-cell dedifferentiation has been revealed as a pathological mechanism underlying pancreatic dysfunction in diabetes. We previously showed that increased miR-7 levels trigger β-cell dedifferentiation and diabetes. We used β-cell-specific miR-7 overexpressing mice (Tg7) to test the hypothesis that loss of β-cell identity triggered by miR-7 overexpression alters islet gene expression and islet microenvironment in diabetes. METHODS We performed bulk and single-cell RNA sequencing (RNA-seq) in islets obtained from β-cell-specific miR-7 overexpressing mice (Tg7). We carried out loss- and gain-of-function experiments in MIN6 and EndoC-bH1 cell lines. We analysed previously published mouse and human T2D data sets. RESULTS Bulk RNA-seq revealed that β-cell dedifferentiation is associated with the induction of genes associated with epithelial-to-mesenchymal transition (EMT) in prediabetic (2-week-old) and diabetic (12-week-old) Tg7 mice. Single-cell RNA-seq (scRNA-seq) indicated that this EMT signature is enriched specifically in β-cells. These molecular changes are associated with a weakening of β-cell: β-cell contacts, increased extracellular matrix (ECM) deposition, and TGFβ-dependent islet fibrosis. We found that the mesenchymal reprogramming of β-cells is explained in part by the downregulation of Pdx1 and its inability to regulate a myriad of epithelial-specific genes expressed in β-cells. Notable among genes transactivated by Pdx1 is Ovol2, which encodes a transcriptional repressor of the EMT transcription factor Zeb2. Following compromised β-cell identity, the reduction in Pdx1 gene expression causes a decrease in Ovol2 protein, triggering mesenchymal reprogramming of β-cells through the induction of Zeb2. We provided evidence that EMT signalling associated with the upregulation of Zeb2 expression is a molecular feature of islets in T2D subjects. CONCLUSIONS Our study indicates that miR-7-mediated β-cell dedifferentiation induces EMT signalling and a chronic response to tissue injury, which alters the islet microenvironment and predisposes to fibrosis. This research suggests that regulators of EMT signalling may represent novel therapeutic targets for treating β-cell dysfunction and fibrosis in T2D.
Collapse
Affiliation(s)
- Daniel S de Jesus
- Cellular Identity and Metabolism Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Tracy C S Mak
- Cellular Identity and Metabolism Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Yi-Fang Wang
- Computing and Bioinformatics Facility, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK
| | - Yorrick von Ohlen
- Cellular Identity and Metabolism Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Ying Bai
- Cellular Identity and Metabolism Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Eva Kane
- Cellular Identity and Metabolism Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | | | - Catherine M Chahrour
- Computing and Bioinformatics Facility, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK
| | | | - Victoria Salem
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern Weg 7, 8093 Zurich, Switzerland
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London W12 0NN, UK; Lee Kong China School of Medicine, Nan Yang Technological University, Singapore
| | - Mathieu Latreille
- Cellular Identity and Metabolism Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
15
|
Hogan JP, Peercy BE. Flipping the switch on the hub cell: Islet desynchronization through cell silencing. PLoS One 2021; 16:e0248974. [PMID: 33831017 PMCID: PMC8031451 DOI: 10.1371/journal.pone.0248974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/08/2021] [Indexed: 11/18/2022] Open
Abstract
Pancreatic β cells, responsible for secreting insulin into the bloodstream and maintaining glucose homeostasis, are organized in the islets of Langerhans as clusters of electrically coupled cells. Gap junctions, connecting neighboring cells, coordinate the behavior of the islet, leading to the synchronized oscillations in the intracellular calcium and insulin secretion in healthy islets. Recent experimental work has shown that silencing special hub cells can lead to a disruption in the coordinated behavior, calling into question the democratic paradigm of islet insulin secretion with more or less equal input from each β cell. Islets were shown to have scale-free functional connectivity and a hub cell whose silencing would lead to a loss of functional connectivity and activity in the islet. A mechanistic model representing the electrical and calcium dynamics of β cells during insulin secretion was applied to a network of cells connected by gap junctions to test the hypothesis of hub cells. Functional connectivity networks were built from the simulated calcium traces, with some networks classified as scale-free, confirming experimental results. Potential hub cells were identified using previously defined centrality measures, but silencing them was unable to desynchronize the islet. Instead, switch cells, which were able to turn off the activity of the islet but were not highly functionally connected, were found via systematically silencing each cell in the network.
Collapse
Affiliation(s)
- Janita P. Hogan
- Department of Mathematics & Statistics, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| | - Bradford E. Peercy
- Department of Mathematics & Statistics, University of Maryland Baltimore County, Baltimore, Maryland, United States of America
| |
Collapse
|
16
|
López–Noriega L, Rutter GA. Long Non-Coding RNAs as Key Modulators of Pancreatic β-Cell Mass and Function. Front Endocrinol (Lausanne) 2021; 11:610213. [PMID: 33628198 PMCID: PMC7897662 DOI: 10.3389/fendo.2020.610213] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
Numerous studies have sought to decipher the genetic and other mechanisms contributing to β-cell loss and dysfunction in diabetes mellitus. However, we have yet to fully understand the etiology of the disease or to develop satisfactory treatments. Since the majority of diabetes susceptibility loci are mapped to non-coding regions within the genome, understanding the functions of non-coding RNAs in β-cell biology might provide crucial insights into the pathogenesis of type 1 (T1D) and type 2 (T2D) diabetes. During the past decade, numerous studies have indicated that long non-coding RNAs play important roles in the maintenance of β-cell mass and function. Indeed, lncRNAs have been shown to be involved in controlling β-cell proliferation during development and/or β-cell compensation in response to hyperglycaemia. LncRNAs such as TUG-1 and MEG3 play a role in both β-cell apoptosis and function, while others sensitize β-cells to apoptosis in response to stress signals. In addition, several long non-coding RNAs have been shown to regulate the expression of β-cell-enriched transcription factors in cis or in trans. In this review, we provide an overview of the roles of lncRNAs in maintaining β-function and mass, and discuss their relevance in the development of diabetes.
Collapse
Affiliation(s)
- Livia López–Noriega
- Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Diabetes, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Diabetes, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
17
|
Chabosseau P, Rutter GA, Millership SJ. Importance of Both Imprinted Genes and Functional Heterogeneity in Pancreatic Beta Cells: Is There a Link? Int J Mol Sci 2021; 22:1000. [PMID: 33498234 PMCID: PMC7863946 DOI: 10.3390/ijms22031000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/02/2023] Open
Abstract
Diabetes mellitus now affects more than 400 million individuals worldwide, with significant impacts on the lives of those affected and associated socio-economic costs. Although defects in insulin secretion underlie all forms of the disease, the molecular mechanisms which drive them are still poorly understood. Subsets of specialised beta cells have, in recent years, been suggested to play critical roles in "pacing" overall islet activity. The molecular nature of these cells, the means through which their identity is established and the changes which may contribute to their functional demise and "loss of influence" in both type 1 and type 2 diabetes are largely unknown. Genomic imprinting involves the selective silencing of one of the two parental alleles through DNA methylation and modified imprinted gene expression is involved in a number of diseases. Loss of expression, or loss of imprinting, can be shown in mouse models to lead to defects in beta cell function and abnormal insulin secretion. In the present review we survey the evidence that altered expression of imprinted genes contribute to loss of beta cell function, the importance of beta cell heterogeneity in normal and disease states, and hypothesise whether there is a direct link between the two.
Collapse
Affiliation(s)
| | | | - Steven J. Millership
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK; (P.C.); (G.A.R.)
| |
Collapse
|
18
|
Patwardhan J, Peercy BE. Network Analysis Applied to Pancreatic Islets. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11469-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
19
|
Pang H, Luo S, Xiao Y, Xia Y, Li X, Huang G, Xie Z, Zhou Z. Emerging Roles of Exosomes in T1DM. Front Immunol 2020; 11:593348. [PMID: 33324409 PMCID: PMC7725901 DOI: 10.3389/fimmu.2020.593348] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/02/2020] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a complex autoimmune disorder that mainly affects children and adolescents. The elevated blood glucose level of patients with T1DM results from absolute insulin deficiency and leads to hyperglycemia and the development of life-threatening diabetic complications. Although great efforts have been made to elucidate the pathogenesis of this disease, the precise underlying mechanisms are still obscure. Emerging evidence indicates that small extracellular vesicles, namely, exosomes, take part in intercellular communication and regulate interorgan crosstalk. More importantly, many findings suggest that exosomes and their cargo are associated with the development of T1DM. Therefore, a deeper understanding of exosomes is beneficial for further elucidating the pathogenic process of T1DM. Exosomes are promising biomarkers for evaluating the risk of developingty T1DM, monitoring the disease state and predicting related complications because their number and composition can reflect the status of their parent cells. Additionally, since exosomes are natural carriers of functional proteins, RNA and DNA, they can be used as therapeutic tools to deliver these molecules and drugs. In this review, we briefly introduce the current understanding of exosomes. Next, we focus on the relationship between exosomes and T1DM from three perspectives, i.e., the pathogenic role of exosomes in T1DM, exosomes as novel biomarkers of T1DM and exosomes as therapeutic tools for T1DM.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Xia
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
20
|
Félix-Martínez GJ, N. Mata A, Godínez-Fernández JR. Reconstructing human pancreatic islet architectures using computational optimization. Islets 2020; 12:121-133. [PMID: 33090076 PMCID: PMC7751670 DOI: 10.1080/19382014.2020.1823178] [Citation(s) in RCA: 5] [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] [Indexed: 12/30/2022] Open
Abstract
We outline a general methodology based on computational optimization and experimental data to reconstruct human pancreatic islet architectures. By using the nuclei coordinates of islet cells obtained through DAPI staining, cell types identified by immunostaining, and cell size distributions estimated from capacitance measurements, reconstructed islets composed of non-overlapping spherical cells were obtained through an iterative optimization procedure. In all cases, the reconstructed architectures included >99% of the experimental identified cells, each of them having a radius within the experimentally reported ranges. Given the wide use of mathematical modeling for the study of pancreatic cells, and recently, of cell-cell interactions within the pancreatic islets, the methodology here proposed, also capable of identifying cell-to-cell contacts, is aimed to provide with a framework for modeling and analyzing experimentally-based pancreatic islet architectures.
Collapse
Affiliation(s)
- Gerardo J. Félix-Martínez
- Cátedras CONACYT, Consejo Nacional de Ciencia y Tecnología, Mexico City, México
- Department of Electrical Engineering, Universidad Autónoma Metropolitana, Mexico City, México
- CONTACT Gerardo J. Félix-Martínez Laboratory of Biophysics AT-221, Universidad Autónoma Metropolitana; San Rafael Atlixco 186, Col. Vicentina, 09340, Iztalapapa, CDMX, México
| | - Aurelio N. Mata
- Department of Electrical Engineering, Universidad Autónoma Metropolitana, Mexico City, México
| | | |
Collapse
|
21
|
Georgiadou E, Haythorne E, Dickerson MT, Lopez-Noriega L, Pullen TJ, da Silva Xavier G, Davis SPX, Martinez-Sanchez A, Semplici F, Rizzuto R, McGinty JA, French PM, Cane MC, Jacobson DA, Leclerc I, Rutter GA. The pore-forming subunit MCU of the mitochondrial Ca 2+ uniporter is required for normal glucose-stimulated insulin secretion in vitro and in vivo in mice. Diabetologia 2020; 63:1368-1381. [PMID: 32350566 PMCID: PMC7286857 DOI: 10.1007/s00125-020-05148-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/27/2020] [Indexed: 12/28/2022]
Abstract
AIMS/HYPOTHESIS Mitochondrial oxidative metabolism is central to glucose-stimulated insulin secretion (GSIS). Whether Ca2+ uptake into pancreatic beta cell mitochondria potentiates or antagonises this process is still a matter of debate. Although the mitochondrial Ca2+ importer (MCU) complex is thought to represent the main route for Ca2+ transport across the inner mitochondrial membrane, its role in beta cells has not previously been examined in vivo. METHODS Here, we inactivated the pore-forming subunit of the MCU, encoded by Mcu, selectively in mouse beta cells using Ins1Cre-mediated recombination. Whole or dissociated pancreatic islets were isolated and used for live beta cell fluorescence imaging of cytosolic or mitochondrial Ca2+ concentration and ATP production in response to increasing glucose concentrations. Electrophysiological recordings were also performed on whole islets. Serum and blood samples were collected to examine oral and i.p. glucose tolerance. RESULTS Glucose-stimulated mitochondrial Ca2+ accumulation (p< 0.05), ATP production (p< 0.05) and insulin secretion (p< 0.01) were strongly inhibited in beta cell-specific Mcu-null (βMcu-KO) animals, in vitro, as compared with wild-type (WT) mice. Interestingly, cytosolic Ca2+ concentrations increased (p< 0.001), whereas mitochondrial membrane depolarisation improved in βMcu-KO animals. βMcu-KO mice displayed impaired in vivo insulin secretion at 5 min (p< 0.001) but not 15 min post-i.p. injection of glucose, whilst the opposite phenomenon was observed following an oral gavage at 5 min. Unexpectedly, glucose tolerance was improved (p< 0.05) in young βMcu-KO (<12 weeks), but not in older animals vs WT mice. CONCLUSIONS/INTERPRETATION MCU is crucial for mitochondrial Ca2+ uptake in pancreatic beta cells and is required for normal GSIS. The apparent compensatory mechanisms that maintain glucose tolerance in βMcu-KO mice remain to be established.
Collapse
Affiliation(s)
- Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Elizabeth Haythorne
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Matthew T Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Livia Lopez-Noriega
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Timothy J Pullen
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Gabriela da Silva Xavier
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Samuel P X Davis
- Photonics Group, Department of Physics, Imperial College London, London, UK
| | - Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Francesca Semplici
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - James A McGinty
- Photonics Group, Department of Physics, Imperial College London, London, UK
| | - Paul M French
- Photonics Group, Department of Physics, Imperial College London, London, UK
| | - Matthew C Cane
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - David A Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
22
|
Carrat GR, Haythorne E, Tomas A, Haataja L, Müller A, Arvan P, Piunti A, Cheng K, Huang M, Pullen TJ, Georgiadou E, Stylianides T, Amirruddin NS, Salem V, Distaso W, Cakebread A, Heesom KJ, Lewis PA, Hodson DJ, Briant LJ, Fung AC, Sessions RB, Alpy F, Kong AP, Benke PI, Torta F, Teo AKK, Leclerc I, Solimena M, Wigley DB, Rutter GA. The type 2 diabetes gene product STARD10 is a phosphoinositide-binding protein that controls insulin secretory granule biogenesis. Mol Metab 2020; 40:101015. [PMID: 32416313 PMCID: PMC7322359 DOI: 10.1016/j.molmet.2020.101015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/24/2020] [Accepted: 05/05/2020] [Indexed: 02/09/2023] Open
Abstract
OBJECTIVE Risk alleles for type 2 diabetes at the STARD10 locus are associated with lowered STARD10 expression in the β-cell, impaired glucose-induced insulin secretion, and decreased circulating proinsulin:insulin ratios. Although likely to serve as a mediator of intracellular lipid transfer, the identity of the transported lipids and thus the pathways through which STARD10 regulates β-cell function are not understood. The aim of this study was to identify the lipids transported and affected by STARD10 in the β-cell and the role of the protein in controlling proinsulin processing and insulin granule biogenesis and maturation. METHODS We used isolated islets from mice deleted selectively in the β-cell for Stard10 (βStard10KO) and performed electron microscopy, pulse-chase, RNA sequencing, and lipidomic analyses. Proteomic analysis of STARD10 binding partners was executed in the INS1 (832/13) cell line. X-ray crystallography followed by molecular docking and lipid overlay assay was performed on purified STARD10 protein. RESULTS βStard10KO islets had a sharply altered dense core granule appearance, with a dramatic increase in the number of "rod-like" dense cores. Correspondingly, basal secretion of proinsulin was increased versus wild-type islets. The solution of the crystal structure of STARD10 to 2.3 Å resolution revealed a binding pocket capable of accommodating polyphosphoinositides, and STARD10 was shown to bind to inositides phosphorylated at the 3' position. Lipidomic analysis of βStard10KO islets demonstrated changes in phosphatidylinositol levels, and the inositol lipid kinase PIP4K2C was identified as a STARD10 binding partner. Also consistent with roles for STARD10 in phosphoinositide signalling, the phosphoinositide-binding proteins Pirt and Synaptotagmin 1 were amongst the differentially expressed genes in βStard10KO islets. CONCLUSION Our data indicate that STARD10 binds to, and may transport, phosphatidylinositides, influencing membrane lipid composition, insulin granule biosynthesis, and insulin processing.
Collapse
Affiliation(s)
- Gaelle R. Carrat
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Elizabeth Haythorne
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andreas Müller
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich, University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany,Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alexandra Piunti
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK,Lille 1 University-Science and Technology, Cité Scientifique, 59655, Villeneuve d'Ascq Cedex, France
| | - Kaiying Cheng
- Section of Structural Biology, Department of Medicine, Imperial College London, London, UK
| | - Mutian Huang
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Timothy J. Pullen
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK,Department of Diabetes, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Eleni Georgiadou
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Theodoros Stylianides
- Loughborough University, Centre of Innovative and Collaborative Construction Engineering, Leicestershire, LE11 3TU, UK
| | - Nur Shabrina Amirruddin
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A∗STAR, Proteos, Singapore, 138673, Singapore,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Victoria Salem
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK,Section of Investigative Medicine, Department of Medicine, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Walter Distaso
- Imperial College Business School, Imperial College London, Exhibition Road, London, SW7 2AZ, UK
| | - Andrew Cakebread
- London Metallomics Facility, King's College London, Strand, London, WC2R 2LS, UK
| | | | | | - David J. Hodson
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK,Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Linford J. Briant
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Annie C.H. Fung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Richard B. Sessions
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Fabien Alpy
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1258, Centre National de la Recherche Scientifique (CNRS), UMR 7104, Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Alice P.S. Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Peter I. Benke
- Singapore Lipidomics Incubator, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Mdical Drive, Singapore, 117596, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Mdical Drive, Singapore, 117596, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), A∗STAR, Proteos, Singapore, 138673, Singapore,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117596, Singapore
| | - Isabelle Leclerc
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK
| | - Michele Solimena
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany,Paul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich, University Hospital Carl Gustav Carus and Faculty of Medicine of the TU Dresden, Dresden, Germany,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany,Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany
| | - Dale B. Wigley
- Section of Structural Biology, Department of Medicine, Imperial College London, London, UK
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Imperial College London, du Cane Road, London, W12 0NN, UK,Corresponding author. +44 (0)20 7594 3340.
| |
Collapse
|
23
|
Khan R, Tomas A, Rutter GA. Effects on pancreatic Beta and other Islet cells of the glucose-dependent insulinotropic polypeptide. Peptides 2020; 125:170201. [PMID: 31751656 DOI: 10.1016/j.peptides.2019.170201] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) is a gut-derived incretin that, in common with glucagon-like peptide-1 (GLP-1), has both insulin releasing and extra-pancreatic glucoregulatory actions. GIP is released in response to glucose or fat absorption and acts on the GIP receptor (GIPR) to potentiate insulin release from pancreatic beta cells. GIP has also been shown to promote beta cell survival and stimulate the release of GLP-1 from islet alpha cells. There is now evidence to suggest that low levels of GIP are secreted from alpha cells and may act in a paracrine manner to prime neighboring beta cells for insulin release. In addition, GIP acts on adipocytes to stimulate fat storage and can exert anorexigenic effects via actions in the hypothalamus. Contrary to GLP-1, the development of effective GIP-based T2D treatments has been hindered by poor bioavailability and attenuation of beta cell responses to GIP in some patients with sub-optimally controlled T2D. Recently, longer-acting GIP agonists that exhibit enzymatic stability, as well as dual GLP-1/GIP agonists which provide simultaneous improvement in glucose and weight control have been generated and successfully tested in animal T2D models. This, together with reports on GIP antagonists that may protect against obesity, has revived the interest on the GIP/GIPR axis as a potential anti-diabetic pathway. In this review, we summarize the known aspects of the effects of GIP on beta and other islet cells and discuss the most recent developments on GIP-based therapeutic agents for the improvement of beta cell function in T2D patients.
Collapse
Affiliation(s)
- Rabeet Khan
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| |
Collapse
|
24
|
Loppini A, Chiodo L. Biophysical modeling of β-cells networks: Realistic architectures and heterogeneity effects. Biophys Chem 2019; 254:106247. [PMID: 31472460 DOI: 10.1016/j.bpc.2019.106247] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/06/2019] [Accepted: 08/06/2019] [Indexed: 11/29/2022]
Abstract
The β-cells dynamics is the regulator of insulin secretion in the pancreas, and its investigation is a central aspect in designing effective treatment strategies for diabetes. Despite great efforts, much is still unknown about the complex organization of such endocrine cells and realistic mathematical modeling represents a useful tool to elucidate key aspects of glucose control in humans. In this contribution, we study the human β-cells collective behaviour, by modeling their electric and metabolic coupling in a cluster, of size and architecture similar to human islets of Langerhans. We focus on the effect of coupling on various dynamics regimes observed in the islets, that are spiking and bursting on multiple timescales. In particular, we test the effect of hubs, that are highly glucose-sensitive β-cells, on the overall network dynamics, observing different modulation depending on the timescale of the dynamics. By properly taking into account the role of cells heterogeneity, recently emerged, our model effectively describes the effect of hubs on the synchronization of the islet response and the correlation of β-cells activity.
Collapse
Affiliation(s)
- A Loppini
- Department of Engineering, University Campus Bio-Medico of Rome, Via Á. del Portillo 21, 00128 Rome, Italy.
| | - L Chiodo
- Department of Engineering, University Campus Bio-Medico of Rome, Via Á. del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
25
|
Salem V, Silva LD, Suba K, Georgiadou E, Neda Mousavy Gharavy S, Akhtar N, Martin-Alonso A, Gaboriau DCA, Rothery SM, Stylianides T, Carrat G, Pullen TJ, Singh SP, Hodson DJ, Leclerc I, Shapiro AMJ, Marchetti P, Briant LJB, Distaso W, Ninov N, Rutter GA. Leader β-cells coordinate Ca 2+ dynamics across pancreatic islets in vivo. Nat Metab 2019; 1:615-629. [PMID: 32694805 PMCID: PMC7617060 DOI: 10.1038/s42255-019-0075-2] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 05/08/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic β-cells form highly connected networks within isolated islets. Whether this behaviour pertains to the situation in vivo, after innervation and during continuous perfusion with blood, is unclear. In the present study, we used the recombinant Ca2+ sensor GCaMP6 to assess glucose-regulated connectivity in living zebrafish Danio rerio, and in murine or human islets transplanted into the anterior eye chamber. In each setting, Ca2+ waves emanated from temporally defined leader β-cells, and three-dimensional connectivity across the islet increased with glucose stimulation. Photoablation of zebrafish leader cells disrupted pan-islet signalling, identifying these as likely pacemakers. Correspondingly, in engrafted mouse islets, connectivity was sustained during prolonged glucose exposure, and super-connected 'hub' cells were identified. Granger causality analysis revealed a controlling role for temporally defined leaders, and transcriptomic analyses revealed a discrete hub cell fingerprint. We thus define a population of regulatory β-cells within coordinated islet networks in vivo. This population may drive Ca2+ dynamics and pulsatile insulin secretion.
Collapse
Affiliation(s)
- Victoria Salem
- Department of Medicine, Imperial College London, London, UK.
| | - Luis Delgadillo Silva
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Kinga Suba
- Department of Medicine, Imperial College London, London, UK
| | | | | | - Nadeem Akhtar
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | | | - David C A Gaboriau
- Facility for Imaging by Light Microscopy, Imperial College London, London, UK
| | - Stephen M Rothery
- Facility for Imaging by Light Microscopy, Imperial College London, London, UK
| | | | - Gaelle Carrat
- Department of Medicine, Imperial College London, London, UK
| | - Timothy J Pullen
- Department of Diabetes, Faculty of Life Science and Medicine, King's College London, London, UK
| | - Sumeet Pal Singh
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Nottingham, UK
| | | | - A M James Shapiro
- Clinical Islet Laboratory and Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | - Nikolay Ninov
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany.
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus of TU Dresden, German Center for Diabetes Research, Dresden, Germany.
| | - Guy A Rutter
- Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
26
|
Navarro-Tableros V, Gomez Y, Brizzi MF, Camussi G. Generation of Human Stem Cell-Derived Pancreatic Organoids (POs) for Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1212:179-220. [PMID: 31025308 DOI: 10.1007/5584_2019_340] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Insulin-dependent diabetes mellitus or type 1 diabetes mellitus (T1DM) is an auto-immune condition characterized by the loss of pancreatic β-cells. The curative approach for highly selected patients is the pancreas or the pancreatic islet transplantation. Nevertheless, these options are limited by a growing shortage of donor organs and by the requirement of immunosuppression.Xenotransplantation of porcine islets has been extensively investigated. Nevertheless, the strong xenoimmunity and the risk of transmission of porcine endogenous retroviruses, have limited their application in clinic. Generation of β-like cells from stem cells is one of the most promising strategies in regenerative medicine. Embryonic, and more recently, adult stem cells are currently the most promising cell sources exploited to generate functional β-cells in vitro. A number of studies demonstrated that stem cells could generate functional pancreatic organoids (POs), able to restore normoglycemia when implanted in different preclinical diabetic models. Nevertheless, a gradual loss of function and cell dead are commonly detected when POs are transplanted in immunocompetent animals. So far, the main issue to be solved is the post-transplanted islet loss, due to the host immune attack. To avoid this hurdle, nanotechnology has provided a number of polymers currently under investigation for islet micro and macro-encapsulation. These new approaches, besides conferring PO immune protection, are able to supply oxygen and nutrients and to preserve PO morphology and long-term viability.Herein, we summarize the current knowledge on bioengineered POs and the stem cell differentiation platforms. We also discuss the in vitro strategies used to generate functional POs, and the protocols currently used to confer immune-protection against the host immune attack (micro- and macro-encapsulation). In addition, the most relevant ongoing clinical trials, and the most relevant hurdles met to move towards clinical application are revised.
Collapse
Affiliation(s)
- Victor Navarro-Tableros
- 2i3T Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico Scarl, University of Turin, Turin, Italy
| | - Yonathan Gomez
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy.
- Fondazione per la Ricerca Biomedica-ONLUS, Turin, Italy.
| |
Collapse
|
27
|
Harata M, Liu S, Promes JA, Burand AJ, Ankrum JA, Imai Y. Delivery of shRNA via lentivirus in human pseudoislets provides a model to test dynamic regulation of insulin secretion and gene function in human islets. Physiol Rep 2018; 6:e13907. [PMID: 30370689 PMCID: PMC6204361 DOI: 10.14814/phy2.13907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 10/01/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022] Open
Abstract
Rodent islets are widely used to study the pathophysiology of beta cells and islet function, however, structural and functional differences exist between human and rodent islets, highlighting the need for human islet studies. Human islets are highly variable, deteriorate during culture, and are difficult to genetically modify, making mechanistic studies difficult to conduct and reproduce. To overcome these limitations, we tested whether pseudoislets, created by dissociation and reaggregation of islet cell suspensions, allow for assessment of dynamic islet function after genetic modulation. Characterization of pseudoislets cultured for 1 week revealed better preservation of first-phase glucose-stimulated insulin secretion (GSIS) compared with cultured-intact islets and insulin secretion profiles similar to fresh islets when challenged by glibenclamide and KCl. qPCR indicated that pseudoislets are similar to the original islets for the expression of markers for cell types, beta cell function, and cellular stress with the exception of reduced proinflammatory cytokine genes (IL1B, CCL2, CXCL8). The expression of extracellular matrix markers (ASPN, COL1A1, COL4A1) was also altered in pseudoislets compared with intact islets. Compared with intact islets transduced by adenovirus, pseudoislets transduced by lentivirus showed uniform transduction and better first-phase GSIS. Lastly, the lentiviral-mediated delivery of short hairpin RNA targeting glucokinase (GCK) achieved significant reduction of GCK expression in pseudoislets as well as marked reduction of both first and second phase GSIS without affecting the insulin secretion in response to KCl. Thus, pseudoislets are a tool that enables efficient genetic modulation of human islet cells while preserving insulin secretion.
Collapse
Affiliation(s)
- Mikako Harata
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIowa
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIowa
| | - Siming Liu
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIowa
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIowa
| | - Joseph A. Promes
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIowa
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIowa
| | - Anthony J. Burand
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIowa
- Department of Biomedical EngineeringUniversity of IowaIowa CityIowa
| | - James A. Ankrum
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIowa
- Department of Biomedical EngineeringUniversity of IowaIowa CityIowa
| | - Yumi Imai
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIowa
- Fraternal Order of Eagles Diabetes Research CenterUniversity of IowaIowa CityIowa
| |
Collapse
|
28
|
Nasteska D, Hodson DJ. The role of beta cell heterogeneity in islet function and insulin release. J Mol Endocrinol 2018; 61:R43-R60. [PMID: 29661799 PMCID: PMC5976077 DOI: 10.1530/jme-18-0011] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/16/2018] [Indexed: 12/15/2022]
Abstract
It is becoming increasingly apparent that not all insulin-secreting beta cells are equal. Subtle differences exist at the transcriptomic and protein expression levels, with repercussions for beta cell survival/proliferation, calcium signalling and insulin release. Notably, beta cell heterogeneity displays plasticity during development, metabolic stress and type 2 diabetes mellitus (T2DM). Thus, heterogeneity or lack thereof may be an important contributor to beta cell failure during T2DM in both rodents and humans. The present review will discuss the molecular and cellular features of beta cell heterogeneity at both the single-cell and islet level, explore how this influences islet function and insulin release and look into the alterations that may occur during obesity and T2DM.
Collapse
Affiliation(s)
- Daniela Nasteska
- Institute of Metabolism and Systems Research (IMSR)University of Birmingham, Edgbaston, UK
- Centre for EndocrinologyDiabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- COMPARE University of Birmingham and University of Nottingham MidlandsBirmingham, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR)University of Birmingham, Edgbaston, UK
- Centre for EndocrinologyDiabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
- COMPARE University of Birmingham and University of Nottingham MidlandsBirmingham, UK
| |
Collapse
|
29
|
Gosak M, Markovič R, Dolenšek J, Slak Rupnik M, Marhl M, Stožer A, Perc M. Network science of biological systems at different scales: A review. Phys Life Rev 2018; 24:118-135. [DOI: 10.1016/j.plrev.2017.11.003] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 12/20/2022]
|
30
|
Gosak M, Stožer A, Markovič R, Dolenšek J, Perc M, Rupnik MS, Marhl M. Critical and Supercritical Spatiotemporal Calcium Dynamics in Beta Cells. Front Physiol 2017; 8:1106. [PMID: 29312008 PMCID: PMC5743929 DOI: 10.3389/fphys.2017.01106] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 12/14/2017] [Indexed: 01/12/2023] Open
Abstract
A coordinated functioning of beta cells within pancreatic islets is mediated by oscillatory membrane depolarization and subsequent changes in cytoplasmic calcium concentration. While gap junctions allow for intraislet information exchange, beta cells within islets form complex syncytia that are intrinsically nonlinear and highly heterogeneous. To study spatiotemporal calcium dynamics within these syncytia, we make use of computational modeling and confocal high-speed functional multicellular imaging. We show that model predictions are in good agreement with experimental data, especially if a high degree of heterogeneity in the intercellular coupling term is assumed. In particular, during the first few minutes after stimulation, the probability distribution of calcium wave sizes is characterized by a power law, thus indicating critical behavior. After this period, the dynamics changes qualitatively such that the number of global intercellular calcium events increases to the point where the behavior becomes supercritical. To better mimic normal in vivo conditions, we compare the described behavior during supraphysiological non-oscillatory stimulation with the behavior during exposure to a slightly lower and oscillatory glucose challenge. In the case of this protocol, we observe only critical behavior in both experiment and model. Our results indicate that the loss of oscillatory changes, along with the rise in plasma glucose observed in diabetes, could be associated with a switch to supercritical calcium dynamics and loss of beta cell functionality.
Collapse
Affiliation(s)
- Marko Gosak
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Andraž Stožer
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Education, University of Maribor, Maribor, Slovenia
- Faculty of Energy Technology, University of Maribor, Krško, Slovenia
| | - Jurij Dolenšek
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
| | - Matjaž Perc
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Center for Applied Mathematics and Theoretical Physics, University of Maribor, Maribor, Slovenia
- Complexity Science Hub, Vienna, Austria
| | - Marjan S. Rupnik
- Faculty of Medicine, Institute of Physiology, University of Maribor, Maribor, Slovenia
- Institute of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
- Faculty of Education, University of Maribor, Maribor, Slovenia
| |
Collapse
|
31
|
Skelin Klemen M, Dolenšek J, Slak Rupnik M, Stožer A. The triggering pathway to insulin secretion: Functional similarities and differences between the human and the mouse β cells and their translational relevance. Islets 2017; 9:109-139. [PMID: 28662366 PMCID: PMC5710702 DOI: 10.1080/19382014.2017.1342022] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In β cells, stimulation by metabolic, hormonal, neuronal, and pharmacological factors is coupled to secretion of insulin through different intracellular signaling pathways. Our knowledge about the molecular machinery supporting these pathways and the patterns of signals it generates comes mostly from rodent models, especially the laboratory mouse. The increased availability of human islets for research during the last few decades has yielded new insights into the specifics in signaling pathways leading to insulin secretion in humans. In this review, we follow the most central triggering pathway to insulin secretion from its very beginning when glucose enters the β cell to the calcium oscillations it produces to trigger fusion of insulin containing granules with the plasma membrane. Along the way, we describe the crucial building blocks that contribute to the flow of information and focus on their functional role in mice and humans and on their translational implications.
Collapse
Affiliation(s)
- Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Institute of Physiology; Center for Physiology and Pharmacology; Medical University of Vienna; Vienna, Austria
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
32
|
Zinman B, Skyler JS, Riddle MC, Ferrannini E. Diabetes Research and Care Through the Ages. Diabetes Care 2017; 40:1302-1313. [PMID: 28931706 DOI: 10.2337/dci17-0042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/26/2017] [Indexed: 02/03/2023]
Abstract
As has been well established, the Diabetes Care journal's most visible signature event is the Diabetes Care Symposium held each year during the American Diabetes Association's Scientific Sessions. Held this past year on 10 June 2017 in San Diego, California, at the 77th Scientific Sessions, this event has become one of the most attended sessions during the Scientific Sessions. Each year, in order to continue to have the symposium generate interest, we revise the format and content of this event. For this past year, our 6th annual symposium, I felt it was time to provide a comprehensive overview of our efforts in diabetes care to determine, first and foremost, how we arrived at our current state of management. I also felt the narrative needed to include the current status of management, especially with a focus toward cardiovascular disease, and finally, we wanted to ask what the future holds. Toward this goal, I asked four of the most noted experts in the world to provide their opinion on this topic. The symposium started with a very thoughtful presentation by Dr. Jay Skyler entitled "A Look Back as to How We Got Here." That was followed by two lectures on current concepts by Dr. Bernard Zinman entitled "Current Treatment Paradigms Today-How Well Are We Doing?" and by Dr. Matthew Riddle entitled "Evolving Concepts and Future Directions for Cardiovascular Outcomes Trials." The final lecture for the symposium was delivered by Dr. Ele Ferrannini and was entitled "What Does the Future Hold?" As always, a well-attended and well-received symposium is now the norm for our signature event and our efforts were rewarded by the enthusiasm of the attendees. This narrative summarizes the lectures held at the symposium.-William T. CefaluChief Scientific, Medical & Mission Officer, American Diabetes Association.
Collapse
Affiliation(s)
- Bernard Zinman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami, Miami, FL
| | - Matthew C Riddle
- Division of Endocrinology, Diabetes & Clinical Nutrition, Oregon Health & Science University, Portland, OR
| | - Ele Ferrannini
- CNR Institute of Clinical Physiology, and the Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
33
|
Makhmutova M, Liang T, Gaisano H, Caicedo A, Almaça J. Confocal Imaging of Neuropeptide Y-pHluorin: A Technique to Visualize Insulin Granule Exocytosis in Intact Murine and Human Islets. J Vis Exp 2017. [PMID: 28930993 DOI: 10.3791/56089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Insulin secretion plays a central role in glucose homeostasis under normal physiological conditions as well as in disease. Current approaches to study insulin granule exocytosis either use electrophysiology or microscopy coupled to the expression of fluorescent reporters. However most of these techniques have been optimized for clonal cell lines or require dissociating pancreatic islets. In contrast, the method presented here allows for real time visualization of insulin granule exocytosis in intact pancreatic islets. In this protocol, we first describe the viral infection of isolated pancreatic islets with adenovirus that encodes a pH-sensitive green fluorescent protein (GFP), pHluorin, coupled to neuropeptide Y (NPY). Second, we describe the confocal imaging of islets five days after viral infection and how to monitor the insulin granule secretion. Briefly, the infected islets are placed on a coverslip on an imaging chamber and imaged under an upright laser-scanning confocal microscope while being continuously perfused with extracellular solution containing various stimuli. Confocal images spanning 50 µm of the islet are acquired as time-lapse recordings using a fast-resonant scanner. The fusion of insulin granules with the plasma membrane can be followed over time. This procedure also allows for testing a battery of stimuli in a single experiment, is compatible with both mouse and human islets, and can be combined with various dyes for functional imaging (e.g., membrane potential or cytosolic calcium dyes).
Collapse
Affiliation(s)
| | - Tao Liang
- Department of Medicine, University of Toronto
| | | | | | | |
Collapse
|
34
|
Abstract
The pancreatic β-cell secretes insulin in response to elevated plasma glucose. This review applies an external bioenergetic critique to the central processes of glucose-stimulated insulin secretion, including glycolytic and mitochondrial metabolism, the cytosolic adenine nucleotide pool, and its interaction with plasma membrane ion channels. The control mechanisms responsible for the unique responsiveness of the cell to glucose availability are discussed from bioenergetic and metabolic control standpoints. The concept of coupling factor facilitation of secretion is critiqued, and an attempt is made to unravel the bioenergetic basis of the oscillatory mechanisms controlling secretion. The need to consider the physiological constraints operating in the intact cell is emphasized throughout. The aim is to provide a coherent pathway through an extensive, complex, and sometimes bewildering literature, particularly for those unfamiliar with the field.
Collapse
Affiliation(s)
- David G Nicholls
- Buck Institute for Research on Aging, Novato, California; and Department of Clinical Sciences, Unit of Molecular Metabolism, Lund University Diabetes Centre, Malmo, Sweden
| |
Collapse
|
35
|
Mitchell RK, Nguyen-Tu MS, Chabosseau P, Callingham RM, Pullen TJ, Cheung R, Leclerc I, Hodson DJ, Rutter GA. The transcription factor Pax6 is required for pancreatic β cell identity, glucose-regulated ATP synthesis, and Ca 2+ dynamics in adult mice. J Biol Chem 2017; 292:8892-8906. [PMID: 28377501 PMCID: PMC5448123 DOI: 10.1074/jbc.m117.784629] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/03/2017] [Indexed: 12/20/2022] Open
Abstract
Heterozygous mutations in the human paired box gene PAX6 lead to impaired glucose tolerance. Although embryonic deletion of the Pax6 gene in mice leads to loss of most pancreatic islet cell types, the functional consequences of Pax6 loss in adults are poorly defined. Here we developed a mouse line in which Pax6 was selectively inactivated in β cells by crossing animals with floxed Pax6 alleles to mice expressing the inducible Pdx1CreERT transgene. Pax6 deficiency, achieved by tamoxifen injection, caused progressive hyperglycemia. Although β cell mass was preserved 8 days post-injection, total insulin content and insulin:chromogranin A immunoreactivity were reduced by ∼60%, and glucose-stimulated insulin secretion was eliminated. RNA sequencing and quantitative real-time PCR analyses revealed that, although the expression of key β cell genes, including Ins2, Slc30a8, MafA, Slc2a2, G6pc2, and Glp1r, was reduced after Pax6 deletion, that of several genes that are usually selectively repressed (“disallowed”) in β cells, including Slc16a1, was increased. Assessed in intact islets, glucose-induced ATP:ADP increases were significantly reduced (p < 0.05) in βPax6KO versus control β cells, and the former displayed attenuated increases in cytosolic Ca2+. Unexpectedly, glucose-induced increases in intercellular connectivity were enhanced after Pax6 deletion, consistent with increases in the expression of the glucose sensor glucokinase, but decreases in that of two transcription factors usually expressed in fully differentiated β-cells, Pdx1 and Nkx6.1, were observed in islet “hub” cells. These results indicate that Pax6 is required for the functional identity of adult β cells. Furthermore, deficiencies in β cell glucose sensing are likely to contribute to defective insulin secretion in human carriers of PAX6 mutations.
Collapse
Affiliation(s)
- Ryan K Mitchell
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Marie-Sophie Nguyen-Tu
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Pauline Chabosseau
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Rebecca M Callingham
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Timothy J Pullen
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Rebecca Cheung
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Isabelle Leclerc
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - David J Hodson
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom, .,the Institute of Metabolism and Systems Research and Centre of Membrane Proteins and Receptors, University of Birmingham, Edgbaston B15 2TT, United Kingdom, and.,the Centre for Endocrinology, Diabetes, and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
| | - Guy A Rutter
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology, and Metabolism, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom,
| |
Collapse
|
36
|
Meda P. Gap junction proteins are key drivers of endocrine function. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:124-140. [PMID: 28284720 DOI: 10.1016/j.bbamem.2017.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/07/2023]
Abstract
It has long been known that the main secretory cells of exocrine and endocrine glands are connected by gap junctions, made by a variety of connexin species that ensure their electrical and metabolic coupling. Experiments in culture systems and animal models have since provided increasing evidence that connexin signaling contributes to control the biosynthesis and release of secretory products, as well as to the life and death of secretory cells. More recently, genetic studies have further provided the first lines of evidence that connexins also control the function of human glands, which are central to the pathogenesis of major endocrine diseases. Here, we summarize the recent information gathered on connexin signaling in these systems, since the last reviews on the topic, with particular regard to the pancreatic beta cells which produce insulin, and the renal cells which produce renin. These cells are keys to the development of various forms of diabetes and hypertension, respectively, and combine to account for the exploding, worldwide prevalence of the metabolic syndrome. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, Switzerland.
| |
Collapse
|
37
|
Abstract
The pulsatility of insulin release is disturbed early in type 2 diabetes, but it is not clear whether specialized pacemaker cells drive islet oscillations. In this issue of Cell Metabolism, Johnston et al. (2016) show that specialized hubs, identified as 1%-10% of beta cells with more active mitochondria and less insulin, synchronize beta cell oscillations.
Collapse
Affiliation(s)
- Jelena Kolic
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - James D Johnson
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
38
|
Frank JA, Yushchenko DA, Hodson DJ, Lipstein N, Nagpal J, Rutter GA, Rhee JS, Gottschalk A, Brose N, Schultz C, Trauner D. Photoswitchable diacylglycerols enable optical control of protein kinase C. Nat Chem Biol 2016; 12:755-62. [PMID: 27454932 PMCID: PMC6101201 DOI: 10.1038/nchembio.2141] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/16/2016] [Indexed: 01/02/2023]
Abstract
Increased levels of the second messenger lipid diacylglycerol (DAG) induce downstream signaling events including the translocation of C1-domain-containing proteins toward the plasma membrane. Here, we introduce three light-sensitive DAGs, termed PhoDAGs, which feature a photoswitchable acyl chain. The PhoDAGs are inactive in the dark and promote the translocation of proteins that feature C1 domains toward the plasma membrane upon a flash of UV-A light. This effect is quickly reversed after the termination of photostimulation or by irradiation with blue light, permitting the generation of oscillation patterns. Both protein kinase C and Munc13 can thus be put under optical control. PhoDAGs control vesicle release in excitable cells, such as mouse pancreatic islets and hippocampal neurons, and modulate synaptic transmission in Caenorhabditis elegans. As such, the PhoDAGs afford an unprecedented degree of spatiotemporal control and are broadly applicable tools to study DAG signaling.
Collapse
Affiliation(s)
- James Allen Frank
- Department of Chemistry and Center for Integrated Protein Science, Ludwig Maximilians University Munich, Munich, Germany
| | - Dmytro A Yushchenko
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - David J Hodson
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
- Institute of Metabolism and Systems Research (IMSR) and Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| | - Noa Lipstein
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Jatin Nagpal
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Institute of Biochemistry, Department for Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, ICTEM, Hammersmith Hospital, London, UK
| | - Jeong-Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Alexander Gottschalk
- Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany
- Institute of Biochemistry, Department for Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Carsten Schultz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Dirk Trauner
- Department of Chemistry and Center for Integrated Protein Science, Ludwig Maximilians University Munich, Munich, Germany
| |
Collapse
|
39
|
|
40
|
Cane MC, Parrington J, Rorsman P, Galione A, Rutter GA. The two pore channel TPC2 is dispensable in pancreatic β-cells for normal Ca²⁺ dynamics and insulin secretion. Cell Calcium 2015; 59:32-40. [PMID: 26769314 PMCID: PMC4751975 DOI: 10.1016/j.ceca.2015.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/08/2015] [Accepted: 12/21/2015] [Indexed: 12/21/2022]
Abstract
Ca(2+) signals are central to the stimulation of insulin secretion from pancreatic β-cells by glucose and other agents, including glucagon-like peptide-1 (GLP-1). Whilst Ca(2+) influx through voltage-gated Ca(2+) channels on the plasma membrane is a key trigger for glucose-stimulated secretion, mobilisation of Ca(2+) from acidic stores has been implicated in the control of more localised Ca(2+) changes and membrane potential. Nicotinic acid adenine dinucleotide phosphate (NAADP), generated in β-cells in response to high glucose, is a potent mobiliser of these stores, and has been proposed to act through two pore channels (TPC1 and TPC2, murine gene names Tpcn1 and Tpcn2). Whilst the role of TPC1 in the control of Ca(2+) mobilisation and insulin secretion was recently confirmed, conflicting data exist for TPC2. Here, we used the selective and efficient deleter strain, Ins1Cre to achieve β-cell selective deletion of the Tpcn2 gene in mice. βTpcn2 KO mice displayed normal intraperitoneal and oral glucose tolerance, and glucose-stimulated Ca(2+) dynamics and insulin secretion from islets were similarly normal. GLP-1-induced Ca(2+) increases involved an increase in oscillation frequency from 4.35 to 4.84 per minute (p=0.04) at 8mM glucose, and this increase was unaffected by the absence of Tpcn2. The current data thus indicate that TPC2 is not absolutely required for normal glucose- or incretin-stimulated insulin secretion from the β-cell. Our findings suggest that TPC1, whose expression tended to increase in Tpcn2 null islets, might be sufficient to support normal Ca(2+) dynamics in response to stimulation by nutrients or incretins.
Collapse
Affiliation(s)
- Matthew C Cane
- Section of Cell Biology and Functional Genomics, Imperial College London, Du Cane Road, W12 0NN London, UK
| | - John Parrington
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, UK
| | - Patrik Rorsman
- The Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Imperial College London, Du Cane Road, W12 0NN London, UK.
| |
Collapse
|
41
|
Gosak M, Markovič R, Fajmut A, Marhl M, Hawlina M, Andjelić S. The Analysis of Intracellular and Intercellular Calcium Signaling in Human Anterior Lens Capsule Epithelial Cells with Regard to Different Types and Stages of the Cataract. PLoS One 2015; 10:e0143781. [PMID: 26636768 PMCID: PMC4670133 DOI: 10.1371/journal.pone.0143781] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/09/2015] [Indexed: 11/17/2022] Open
Abstract
In this work we investigated how modifications of the Ca2+ homeostasis in anterior lens epithelial cells (LECs) are associated with different types of cataract (cortical or nuclear) and how the progression of the cataract (mild or moderate) affects the Ca2+ signaling. We systematically analyzed different aspects of intra- and inter-cellular Ca2+ signaling in the human LECs, which are attached to surgically isolated lens capsule (LC), obtained during cataract surgery. We monitored the temporal and spatial changes in intracellular Ca2+ concentration after stimulation with acetylcholine by means of Fura-2 fluorescence captured with an inverted microscope. In our analysis we compared the features of Ca2+ signals in individual cells, synchronized activations, spatio-temporal grouping and the nature of intercellular communication between LECs. The latter was assessed by using the methodologies of the complex network theory. Our results point out that at the level of individual cells there are no significant differences when comparing the features of the signals with regard either to the type or the stage of the cataract. On the other hand, noticeable differences are observed at the multicellular level, despite inter-capsule variability. LCs associated with more developed cataracts were found to exhibit a slower collective response to stimulation, a less pronounced spatio-temporal clustering of LECs with similar signaling characteristics. The reconstructed intercellular networks were found to be sparser and more segregated than in LCs associated with mild cataracts. Moreover, we show that spontaneously active LECs often operate in localized groups with quite well aligned Ca2+ activity. The presence of spontaneous activity was also found to affect the stimulated Ca2+ responses of individual cells. Our findings indicate that the cataract progression entails the impairment of intercellular signaling thereby suggesting the functional importance of altered Ca2+ signaling of LECs in cataractogenesis.
Collapse
Affiliation(s)
- Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, Slovenia.,Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Education, University of Maribor, Maribor, Slovenia
| | - Aleš Fajmut
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Health Sciences, University of Maribor, Maribor, Slovenia
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, Maribor, Slovenia.,Faculty of Education, University of Maribor, Maribor, Slovenia
| | - Marko Hawlina
- Eye Hospital, University Medical Centre, Ljubljana, Slovenia
| | - Sofija Andjelić
- Eye Hospital, University Medical Centre, Ljubljana, Slovenia
| |
Collapse
|
42
|
Dolenšek J, Špelič D, Skelin Klemen M, Žalik B, Gosak M, Slak Rupnik M, Stožer A. Membrane Potential and Calcium Dynamics in Beta Cells from Mouse Pancreas Tissue Slices: Theory, Experimentation, and Analysis. SENSORS 2015; 15:27393-419. [PMID: 26516866 PMCID: PMC4701238 DOI: 10.3390/s151127393] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/11/2015] [Accepted: 10/14/2015] [Indexed: 12/17/2022]
Abstract
Beta cells in the pancreatic islets of Langerhans are precise biological sensors for glucose and play a central role in balancing the organism between catabolic and anabolic needs. A hallmark of the beta cell response to glucose are oscillatory changes of membrane potential that are tightly coupled with oscillatory changes in intracellular calcium concentration which, in turn, elicit oscillations of insulin secretion. Both membrane potential and calcium changes spread from one beta cell to the other in a wave-like manner. In order to assess the properties of the abovementioned responses to physiological and pathological stimuli, the main challenge remains how to effectively measure membrane potential and calcium changes at the same time with high spatial and temporal resolution, and also in as many cells as possible. To date, the most wide-spread approach has employed the electrophysiological patch-clamp method to monitor membrane potential changes. Inherently, this technique has many advantages, such as a direct contact with the cell and a high temporal resolution. However, it allows one to assess information from a single cell only. In some instances, this technique has been used in conjunction with CCD camera-based imaging, offering the opportunity to simultaneously monitor membrane potential and calcium changes, but not in the same cells and not with a reliable cellular or subcellular spatial resolution. Recently, a novel family of highly-sensitive membrane potential reporter dyes in combination with high temporal and spatial confocal calcium imaging allows for simultaneously detecting membrane potential and calcium changes in many cells at a time. Since the signals yielded from both types of reporter dyes are inherently noisy, we have developed complex methods of data denoising that permit for visualization and pixel-wise analysis of signals. Combining the experimental approach of high-resolution imaging with the advanced analysis of noisy data enables novel physiological insights and reassessment of current concepts in unprecedented detail.
Collapse
Affiliation(s)
- Jurij Dolenšek
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
| | - Denis Špelič
- Faculty of Electrical Engineering and Computer Science, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (D.Š.); (B.Ž.)
| | - Maša Skelin Klemen
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
| | - Borut Žalik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (D.Š.); (B.Ž.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
| | - Marko Gosak
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
- Department of Physics, Faculty of Natural Sciences and Mathematics, University of Maribor, SI-2000 Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
- Center for Physiology and Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Andraž Stožer
- Institute of Physiology, Faculty of Medicine, University of Maribor, SI-2000 Maribor, Slovenia; E-Mails: (J.D.); (M.S.K.); (M.G.); (M.S.R.)
- Center for Open Innovation and Research, Core@UM, University of Maribor, SI-2000 Maribor, Slovenia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +386-2-2345843
| |
Collapse
|
43
|
Solomou A, Meur G, Bellomo E, Hodson DJ, Tomas A, Li SM, Philippe E, Herrera PL, Magnan C, Rutter GA. The Zinc Transporter Slc30a8/ZnT8 Is Required in a Subpopulation of Pancreatic α-Cells for Hypoglycemia-induced Glucagon Secretion. J Biol Chem 2015; 290:21432-42. [PMID: 26178371 PMCID: PMC4571871 DOI: 10.1074/jbc.m115.645291] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Indexed: 12/02/2022] Open
Abstract
SLC30A8 encodes a zinc transporter ZnT8 largely restricted to pancreatic islet β- and α-cells, and responsible for zinc accumulation into secretory granules. Although common SLC30A8 variants, believed to reduce ZnT8 activity, increase type 2 diabetes risk in humans, rare inactivating mutations are protective. To investigate the role of Slc30a8 in the control of glucagon secretion, Slc30a8 was inactivated selectively in α-cells by crossing mice with alleles floxed at exon 1 to animals expressing Cre recombinase under the pre-proglucagon promoter. Further crossing to Rosa26:tdRFP mice, and sorting of RFP+: glucagon+ cells from KO mice, revealed recombination in ∼30% of α-cells, of which ∼50% were ZnT8-negative (14 ± 1.8% of all α-cells). Although glucose and insulin tolerance were normal, female αZnT8KO mice required lower glucose infusion rates during hypoglycemic clamps and displayed enhanced glucagon release (p < 0.001) versus WT mice. Correspondingly, islets isolated from αZnT8KO mice secreted more glucagon at 1 mm glucose, but not 17 mm glucose, than WT controls (n = 5; p = 0.008). Although the expression of other ZnT family members was unchanged, cytoplasmic (n = 4 mice per genotype; p < 0.0001) and granular (n = 3, p < 0.01) free Zn2+ levels were significantly lower in KO α-cells versus control cells. In response to low glucose, the amplitude and frequency of intracellular Ca2+ increases were unchanged in α-cells of αZnT8KO KO mice. ZnT8 is thus important in a subset of α-cells for normal responses to hypoglycemia and acts via Ca2+-independent mechanisms.
Collapse
Affiliation(s)
- Antonia Solomou
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Gargi Meur
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Elisa Bellomo
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - David J Hodson
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Alejandra Tomas
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom, the Department of Cell Biology, Institute of Ophthalmology, University College London, Greater London EC1V 9EL, United Kingdom
| | - Stéphanie Migrenne Li
- the University Paris Diderot-Paris 7, Unit of Functional and Adaptive Biology (BFA) EAC 7059 CNRS, 75013 Paris, France, and
| | - Erwann Philippe
- the University Paris Diderot-Paris 7, Unit of Functional and Adaptive Biology (BFA) EAC 7059 CNRS, 75013 Paris, France, and
| | - Pedro L Herrera
- the Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, 1211 Geneva-4, Switzerland
| | - Christophe Magnan
- the University Paris Diderot-Paris 7, Unit of Functional and Adaptive Biology (BFA) EAC 7059 CNRS, 75013 Paris, France, and
| | - Guy A Rutter
- From the Section of Cell Biology and Functional Genomics, Division of Diabetes Endocrinology and Metabolism, Department of Medicine, Imperial College London, London W12 0NN, United Kingdom,
| |
Collapse
|
44
|
Soedling H, Hodson DJ, Adrianssens AE, Gribble FM, Reimann F, Trapp S, Rutter GA. Limited impact on glucose homeostasis of leptin receptor deletion from insulin- or proglucagon-expressing cells. Mol Metab 2015; 4:619-30. [PMID: 26413468 PMCID: PMC4563029 DOI: 10.1016/j.molmet.2015.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/12/2015] [Indexed: 01/07/2023] Open
Abstract
AIMS/HYPOTHESIS The adipose tissue-derived hormone leptin plays an important role in the maintenance of body weight and glucose homeostasis. Leptin mediates its effects by interaction with leptin receptors (LepRb), which are highly expressed in the hypothalamus and other brain centres, and at lower levels in the periphery. Previous studies have used relatively promiscuous or inefficient Cre deleter strains, respectively, to explore the roles of LepR in pancreatic β and α cells. Here, we use two newly-developed Cre lines to explore the role of leptin signalling in insulin and proglucagon-expressing cells. METHODS Leptin receptor expression was measured in isolated mouse islets and highly-purified islet cells by RNASeq and quantitative RT-PCR. Mice lacking leptin signalling in pancreatic β, or in α and other proglucagon-expressing cells, were generated using Ins1Cre- or iGluCre-mediated recombination respectively of flox'd leptin receptor alleles. In vivo glucose homeostasis, changes in body weight, pancreatic histology and hormone secretion from isolated islets were assessed using standard techniques. RESULTS Leptin receptor mRNA levels were at or below the level of detection in wild-type adult mouse isolated islets and purified cells, and leptin signalling to Stat3 phosphorylation was undetectable. Whereas male mice further deleted for leptin receptors in β cells exhibited no abnormalities in glucose tolerance up to 16 weeks of age, females transiently displayed improved glucose tolerance at 8 weeks (11.2 ± 3.2% decrease in area under curve; p < 0.05), and improved (39.0 ± 13.0%, P < 0.05) glucose-stimulated insulin secretion in vitro. No differences were seen between genotypes in body weight, fasting glucose or β/α cell ratio. Deletion of LepR from α-cells, a minority of β cells, and a subset of proglucagon-expressing cells in the brain, exerted no effects on body weight, glucose or insulin tolerance, nor on pancreatic hormone secretion assessed in vivo and in vitro. CONCLUSIONS/INTERPRETATION The use here of a highly selective Cre recombinase indicates that leptin signalling plays a relatively minor, age- and sex-dependent role in the control of β cell function in the mouse. No in vivo role for leptin receptors on α cells, nor in other proglucagon-expressing cells, was detected in this study.
Collapse
Affiliation(s)
- Helen Soedling
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, du Cane Road, London W12 0NN, UK
| | - David J Hodson
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, du Cane Road, London W12 0NN, UK
| | | | - Fiona M Gribble
- University of Cambridge Metabolic Research Laboratories, Cambridge, UK
| | - Frank Reimann
- University of Cambridge Metabolic Research Laboratories, Cambridge, UK
| | - Stefan Trapp
- Centre for Cardiovascular and Metabolic Neuroscience, Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, du Cane Road, London W12 0NN, UK
| |
Collapse
|
45
|
Guay C, Menoud V, Rome S, Regazzi R. Horizontal transfer of exosomal microRNAs transduce apoptotic signals between pancreatic beta-cells. Cell Commun Signal 2015; 13:17. [PMID: 25880779 PMCID: PMC4371845 DOI: 10.1186/s12964-015-0097-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
Background Diabetes mellitus is a common metabolic disorder characterized by dysfunction of insulin-secreting pancreatic beta-cells. MicroRNAs are important regulators of beta-cell activities. These non-coding RNAs have recently been discovered to exert their effects not only inside the cell producing them but, upon exosome-mediated transfer, also in other recipient cells. This novel communication mode remains unexplored in pancreatic beta-cells. In the present study, the microRNA content of exosomes released by beta-cells in physiological and physiopathological conditions was analyzed and the biological impact of their transfer to recipient cells investigated. Results Exosomes were isolated from the culture media of MIN6B1 and INS-1 derived 832/13 beta-cell lines and from mice, rat or human islets. Global profiling revealed that the microRNAs released in MIN6B1 exosomes do not simply reflect the content of the cells of origin. Indeed, while a subset of microRNAs was preferentially released in exosomes others were selectively retained in the cells. Moreover, exposure of MIN6B1 cells to inflammatory cytokines changed the release of several microRNAs. The dynamics of microRNA secretion and their potential transfer to recipient cells were next investigated. As a proof-of-concept, we demonstrate that if cel-miR-238, a C. Elegans microRNA not present in mammalian cells, is expressed in MIN6B1 cells a fraction of it is released in exosomes and is transferred to recipient beta-cells. Furthermore, incubation of untreated MIN6B1 or mice islet cells in the presence of microRNA-containing exosomes isolated from the culture media of cytokine-treated MIN6B1 cells triggers apoptosis of recipient cells. In contrast, exosomes originating from cells not exposed to cytokines have no impact on cell survival. Apoptosis induced by exosomes produced by cytokine-treated cells was prevented by down-regulation of the microRNA-mediating silencing protein Ago2 in recipient cells, suggesting that the effect is mediated by the non-coding RNAs. Conclusions Taken together, our results suggest that beta-cells secrete microRNAs that can be transferred to neighboring beta-cells. Exposure of donor cells to pathophysiological conditions commonly associated with diabetes modifies the release of microRNAs and affects survival of recipient beta-cells. Our results support the concept that exosomal microRNAs transfer constitutes a novel cell-to-cell communication mechanism regulating the activity of pancreatic beta-cells. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0097-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| | - Véronique Menoud
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| | - Sophie Rome
- CarMeN Laboratory (INSERM U.1060/INRA 1397, INSA), University of Lyon, Faculty of Medicine Lyon-Sud, Ouillons, France.
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| |
Collapse
|
46
|
Broichhagen J, Frank JA, Johnston NR, Mitchell RK, Šmid K, Marchetti P, Bugliani M, Rutter GA, Trauner D, Hodson DJ. A red-shifted photochromic sulfonylurea for the remote control of pancreatic beta cell function. Chem Commun (Camb) 2015; 51:6018-21. [PMID: 25744824 DOI: 10.1039/c5cc01224d] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Azobenzene photoresponsive elements can be installed on sulfonylureas, yielding optical control over pancreatic beta cell function and insulin release. An obstacle to such photopharmacological approaches remains the use of ultraviolet-blue illumination. Herein, we synthesize and test a novel yellow light-activated sulfonylurea based on a heterocyclic azobenzene bearing a push-pull system.
Collapse
Affiliation(s)
- J Broichhagen
- Department of Chemistry and Center for Integrated Protein Science, LMU Munich, Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|