1
|
Vyakaranam AR, Mahamed MM, Hellman P, Eriksson O, Espes D, Christoffersson G, Sundin A. Non-invasive imaging of sympathetic innervation of the pancreas in individuals with type 2 diabetes. Diabetologia 2024; 67:199-208. [PMID: 37935826 PMCID: PMC10709256 DOI: 10.1007/s00125-023-06039-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/31/2023] [Indexed: 11/09/2023]
Abstract
AIMS/HYPOTHESIS Compromised pancreatic sympathetic innervation has been suggested as a factor involved in both immune-mediated beta cell destruction and endocrine dysregulation of pancreatic islets. To further explore these intriguing findings, new techniques for in vivo assessment of pancreatic innervation are required. This is a retrospective study that aimed to investigate whether the noradrenaline (norepinephrine) analogue 11C-hydroxy ephedrine (11C-HED) could be used for quantitative positron emission tomography (PET) imaging of the sympathetic innervation of the human pancreas. METHODS In 25 individuals with type 2 diabetes and 64 individuals without diabetes, all of whom had previously undergone 11C-HED-PET/CT because of pheochromocytoma or paraganglioma (or suspicion thereof), the 11C-HED standardised uptake value (SUVmean), 11C-HED specific binding index (SBI), pancreatic functional volume (FV, in ml), functional neuronal volume (FNV, calculated as SUVmean × FV), specific binding index with functional volume (SBI FV, calculated as SBI × FV) and attenuation on CT (HU) were investigated in the entire pancreas, and additionally in six separate anatomical pancreatic regions. RESULTS Generally, 11C-HED uptake in the pancreas was high, with marked individual variation, suggesting variability in sympathetic innervation. Moreover, pancreatic CT attenuation (HU) (p<0.001), 11C-HED SBI (p=0.0049) and SBI FV (p=0.0142) were lower in individuals with type 2 diabetes than in individuals without diabetes, whereas 11C-HED SUVmean (p=0.15), FV (p=0.73) and FNV (p=0.30) were similar. CONCLUSIONS/INTERPRETATION We demonstrate the feasibility of using 11C-HED-PET for non-invasive assessment of pancreatic sympathetic innervation in humans. These findings warrant further prospective evaluation, especially in individuals with theoretical defects in pancreatic sympathetic innervation, such as those with type 1 diabetes.
Collapse
Affiliation(s)
- Achyut Ram Vyakaranam
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden.
| | - Maryama M Mahamed
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Daniel Espes
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gustaf Christoffersson
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Anders Sundin
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023; 325:C141-C154. [PMID: 37273237 PMCID: PMC10312332 DOI: 10.1152/ajpcell.00533.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Caleb J Worker
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Yumei Feng Earley
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| |
Collapse
|
3
|
Zhao Y, Veysman B. Revisiting the Pathogenesis of Type 1 Diabetes: Importance of Neural Input to Pancreatic Islets and the Therapeutic Capability of Stem Cell Educator TM Therapy to Restore Their Integrity. Biomedicines 2023; 11:594. [PMID: 36831130 PMCID: PMC9952924 DOI: 10.3390/biomedicines11020594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease with a shortage of islet β cells. To date, the etiology of T1D remains elusive. Increasing clinical evidence and animal studies demonstrate that autoimmune cells are directed against the nervous system of pancreatic islets, contributing to the development of T1D. Therefore, it highlights the necessity to explore novel clinical approaches to fundamentally correct the T1D autoimmunity not only focusing on islet β cells but also on protecting the islet nervous system. This allows the restoration of the integrity of islet innervation and the normal islet β-cell function. To address these issues, we developed a novel technology designated the Stem Cell Educator TM therapy, based on immune education by human cord-blood-derived multipotent stem cells (CB-SC). International amulticenter clinical trials demonstrated its clinical safety and efficacy to treat T1D and other autoimmune diseases. Stem Cell Educator TM therapy may have the potential to revolutionize the treatment of T1D, without the safety and ethical concerns associated with conventional immune and/or stem cell-based therapies.
Collapse
Affiliation(s)
- Yong Zhao
- Throne Biotechnologies, Paramus, NJ 07652, USA
| | | |
Collapse
|
4
|
Gola M, Sejda A, Godlewski J, Cieślak M, Starzyńska A. Neural Component of the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:5246. [PMID: 36358664 PMCID: PMC9657005 DOI: 10.3390/cancers14215246] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/25/2022] [Indexed: 10/15/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive primary malignancy of the pancreas, with a dismal prognosis and limited treatment options. It possesses a unique tumor microenvironment (TME), generating dense stroma with complex elements cross-talking with each other to promote tumor growth and progression. Diversified neural components makes for not having a full understanding of their influence on its aggressive behavior. The aim of the study was to summarize and integrate the role of nerves in the pancreatic tumor microenvironment. The role of autonomic nerve fibers on PDAC development has been recently studied, which resulted in considering the targeting of sympathetic and parasympathetic pathways as a novel treatment opportunity. Perineural invasion (PNI) is commonly found in PDAC. As the severity of the PNI correlates with a poorer prognosis, new quantification of this phenomenon, distinguishing between perineural and endoneural invasion, could feature in routine pathological examination. The concepts of cancer-related neurogenesis and axonogenesis in PDAC are understudied; so, further research in this field may be warranted. A better understanding of the interdependence between the neural component and cancer cells in the PDAC microenvironment could bring new nerve-oriented treatment options into clinical practice and improve outcomes in patients with pancreatic cancer. In this review, we aim to summarize and integrate the current state of knowledge and future challenges concerning nerve-cancer interactions in PDAC.
Collapse
Affiliation(s)
- Michał Gola
- Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Aleksandra Sejda
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 18 Żołnierska Street, 10-561 Olsztyn, Poland
| | - Janusz Godlewski
- Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Małgorzata Cieślak
- Department of Pathomorphology and Forensic Medicine, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 18 Żołnierska Street, 10-561 Olsztyn, Poland
| | - Anna Starzyńska
- Department of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland
| |
Collapse
|
5
|
Hampton RF, Jimenez-Gonzalez M, Stanley SA. Unravelling innervation of pancreatic islets. Diabetologia 2022; 65:1069-1084. [PMID: 35348820 PMCID: PMC9205575 DOI: 10.1007/s00125-022-05691-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/08/2022] [Indexed: 01/05/2023]
Abstract
The central and peripheral nervous systems play critical roles in regulating pancreatic islet function and glucose metabolism. Over the last century, in vitro and in vivo studies along with examination of human pancreas samples have revealed the structure of islet innervation, investigated the contribution of sympathetic, parasympathetic and sensory neural pathways to glucose control, and begun to determine how the structure and function of pancreatic nerves are disrupted in metabolic disease. Now, state-of-the art techniques such as 3D imaging of pancreatic innervation and targeted in vivo neuromodulation provide further insights into the anatomy and physiological roles of islet innervation. Here, we provide a summary of the published work on the anatomy of pancreatic islet innervation, its roles, and evidence for disordered islet innervation in metabolic disease. Finally, we discuss the possibilities offered by new technologies to increase our knowledge of islet innervation and its contributions to metabolic regulation.
Collapse
Affiliation(s)
- Rollie F Hampton
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Jimenez-Gonzalez
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
6
|
Krivova YS, Proshchina AE, Otlyga DA, Leonova OG, Saveliev SV. Prenatal development of sympathetic innervation of the human pancreas. Ann Anat 2021; 240:151880. [PMID: 34896557 DOI: 10.1016/j.aanat.2021.151880] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The sympathetic nervous system plays an important role in the regulation of pancreatic exocrine and endocrine secretion. The results of experimental studies also demonstrate the involvement of the sympathetic nervous system in the regulation of endocrine cell differentiation and islet formation during the development of the pancreas. However, the prenatal development of sympathetic innervation of the human pancreas has not yet been studied. MATERIAL AND METHODS Pancreatic autopsy samples from 24 human fetuses were examined using immunohistochemistry with antibodies to tyrosine hydroxylase (TH). The density, concentration, and size (width, length, perimeter and area) of the TH-positive sympathetic nerves were compared in four developmental periods: pre-fetal (8-11 weeks post conception (w.p.c.), n = 6), early fetal (13-20 gestational weeks (g.w.), n = 7), middle fetal (21-28 g.w., n = 6) and late fetal (29-40 g.w., n = 5) using morphometric methods and statistical analysis (Multiple Comparisons p values). Double immunofluorescence with antibodies to TH and either insulin or glucagon and confocal microscopy were applied to analyze the interaction between the sympathetic nerves and endocrine cells, and the co-localization of TH with hormones. RESULTS TH-positive sympathetic nerves were detected in the fetal pancreas starting from the early stages (8 w.p.c.). The developmental dynamics of sympathetic nerves was follows: from the pre-fetal period, the amount of TH-positive nerves gradually increased and their branching occurred reaching the highest density and concentration in the middle fetal period, followed by a decrease in these parameters in the late fetal period. From the 14th g.w. onwards, thin TH-positive nerve fibers were mainly distributed in the vicinity of blood vessels and around the neurons of intrapancreatic ganglia, which is similar in adults. There were only rare TH-positive nerve fibers adjacent to acini or located at the periphery of some islets. The close interactions between the TH-positive nerve fibers and endocrine cells were observed in the neuro-insular complexes. Additionally, non-neuronal TH-containing cells were found in the pancreas of fetuses from the pre-fetal and early fetal periods. Some of these cells simultaneously contained glucagon. CONCLUSIONS The results demonstrate that sympathetic innervation of the human pancreas, including the formation of perivascular and intraganglionic nerve plexuses, extensively develops during prenatal period, while some processes, such as the formation of sympathetic innervation of islet capillaries, may occur postnatally. Non-neuronal TH-containing cells, as well as the interactions between the sympathetic terminals and endocrine cells observed in the fetal pancreas may be necessary for endocrine pancreas development in humans.
Collapse
Affiliation(s)
- Yuliya S Krivova
- Research Institute of Human Morphology, Tsurupy st., 3, 117418 Moscow, Russia.
| | | | - Dmitry A Otlyga
- Research Institute of Human Morphology, Tsurupy st., 3, 117418 Moscow, Russia.
| | - Ol'ga G Leonova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova St. 32, 119991 Moscow, Russia.
| | - Sergey V Saveliev
- Research Institute of Human Morphology, Tsurupy st., 3, 117418 Moscow, Russia.
| |
Collapse
|
7
|
Sluga N, Postić S, Sarikas S, Huang YC, Stožer A, Slak Rupnik M. Dual Mode of Action of Acetylcholine on Cytosolic Calcium Oscillations in Pancreatic Beta and Acinar Cells In Situ. Cells 2021; 10:1580. [PMID: 34201461 PMCID: PMC8305080 DOI: 10.3390/cells10071580] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/19/2021] [Indexed: 12/13/2022] Open
Abstract
Cholinergic innervation in the pancreas controls both the release of digestive enzymes to support the intestinal digestion and absorption, as well as insulin release to promote nutrient use in the cells of the body. The effects of muscarinic receptor stimulation are described in detail for endocrine beta cells and exocrine acinar cells separately. Here we describe morphological and functional criteria to separate these two cell types in situ in tissue slices and simultaneously measure their response to ACh stimulation on cytosolic Ca2+ oscillations [Ca2+]c in stimulatory glucose conditions. Our results show that both cell types respond to glucose directly in the concentration range compatible with the glucose transporters they express. The physiological ACh concentration increases the frequency of glucose stimulated [Ca2+]c oscillations in both cell types and synchronizes [Ca2+]c oscillations in acinar cells. The supraphysiological ACh concentration further increases the oscillation frequency on the level of individual beta cells, inhibits the synchronization between these cells, and abolishes oscillatory activity in acinar cells. We discuss possible mechanisms leading to the observed phenomena.
Collapse
Affiliation(s)
- Nastja Sluga
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (N.S.); (A.S.)
| | - Sandra Postić
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
| | - Srdjan Sarikas
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
| | - Ya-Chi Huang
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
| | - Andraž Stožer
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (N.S.); (A.S.)
| | - Marjan Slak Rupnik
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia; (N.S.); (A.S.)
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (S.P.); (S.S.); (Y.-C.H.)
- Alma Mater Europaea, European Center Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
8
|
Cinti F, Mezza T, Severi I, Suleiman M, Cefalo CMA, Sorice GP, Moffa S, Impronta F, Quero G, Alfieri S, Mari A, Pontecorvi A, Marselli L, Cinti S, Marchetti P, Giaccari A. Noradrenergic fibers are associated with beta-cell dedifferentiation and impaired beta-cell function in humans. Metabolism 2021; 114:154414. [PMID: 33129839 DOI: 10.1016/j.metabol.2020.154414] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes (T2D) is characterized by a progressive loss of beta-cell function, and the "disappearance" of beta-cells in T2D may also be caused by the process of beta -cell dedifferentiation. Since noradrenergic innervation inhibits insulin secretion and density of noradrenergic fibers is increased in type 2 diabetes mouse models, we aimed to study the relation between islet innervation, dedifferentiation and beta-cell function in humans. METHODS Using immunohistochemistry and electron microscopy, we analyzed pancreata from organ donors and from patients undergoing pancreatic surgery. In the latter, a pre-surgical detailed metabolic characterization by oral glucose tolerance test (OGTT) and hyperglycemic clamp was performed before surgery, thus obtaining in vivo functional parameters of beta-cell function and insulin secretion. RESULTS The islets of diabetic subjects were 3 times more innervated than controls (0.91 ± 0.21 vs 0.32 ± 0.10, n.fibers/islet; p = 0.01), and directly correlated with the dedifferentiation score (r = 0.39; p = 0.03). In vivo functional parameters of insulin secretion, assessed by hyperglycemic clamp, negatively correlated with the increase in fibers [beta-cell Glucose Sensitivity (r = -0.84; p = 0.01), incremental second-phase insulin secretion (r = -0.84, p = 0.03) and arginine-stimulated insulin secretion (r = -0.76, p = 0.04)]. Moreover, we observed a progressive increase in fibers, paralleling worsening glucose tolerance (from NGT through IGT to T2D). CONCLUSIONS/INTERPRETATION Noradrenergic fibers are significantly increased in the islets of diabetic subjects and this positively correlates with beta-cell dedifferentiation score. The correlation between in vivo insulin secretion parameters and the density of pancreatic noradrenergic fibers suggests a significant involvement of these fibers in the pathogenesis of the disease, and indirectly, in the islet dedifferentiation process.
Collapse
Affiliation(s)
- F Cinti
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - T Mezza
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - I Severi
- Department of Clinical and Experimental Medicine, Center of Obesity, Università Politecnica delle Marche, Ancona, Italy
| | - M Suleiman
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - C M A Cefalo
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - G P Sorice
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - S Moffa
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - F Impronta
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - G Quero
- Chirurgia Digestiva, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Istituto di Semeiotica Chirurgica, Università Cattolica del Sacro Cuore, Roma, Italy
| | - S Alfieri
- Chirurgia Digestiva, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Istituto di Semeiotica Chirurgica, Università Cattolica del Sacro Cuore, Roma, Italy
| | - A Mari
- Institute of Neuroscience, National Research Council, Padua, Italy
| | - A Pontecorvi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - L Marselli
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - S Cinti
- Department of Clinical and Experimental Medicine, Center of Obesity, Università Politecnica delle Marche, Ancona, Italy
| | - P Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - A Giaccari
- Centro per le Malattie Endocrine e Metaboliche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| |
Collapse
|
9
|
Abstract
At the time of Ivan Pavlov, pancreatic innervation was studied by looking at pancreas secretions in response to electrical stimulation of nerves. Nowadays we have ways to visualize neuronal activity in real time thanks to advances in fluorescent reporters and imaging techniques. We also have very precise optogenetic and pharmacogenetic approaches that allow neuronal manipulations in a very specific manner. These technological advances have been extensively employed for studying the central nervous system and are just beginning to be incorporated for studying visceral innervation. Pancreatic innervation is complex, and the role it plays in physiology and pathophysiology of the organ is still not fully understood. In this review we highlight anatomical aspects of pancreatic innervation, techniques for pancreatic neuronal labeling, and approaches for imaging pancreatic innervation in vitro and in vivo.
Collapse
|
10
|
Akolpoglu MB, Inceoglu Y, Bozuyuk U, Sousa AR, Oliveira MB, Mano JF, Kizilel S. Recent advances in the design of implantable insulin secreting heterocellular islet organoids. Biomaterials 2020; 269:120627. [PMID: 33401104 DOI: 10.1016/j.biomaterials.2020.120627] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Islet transplantation has proved one of the most remarkable transmissions from an experimental curiosity into a routine clinical application for the treatment of type I diabetes (T1D). Current efforts for taking this technology one-step further are now focusing on overcoming islet donor shortage, engraftment, prolonged islet availability, post-transplant vascularization, and coming up with new strategies to eliminate lifelong immunosuppression. To this end, insulin secreting 3D cell clusters composed of different types of cells, also referred as heterocellular islet organoids, spheroids, or pseudoislets, have been engineered to overcome the challenges encountered by the current islet transplantation protocols. β-cells or native islets are accompanied by helper cells, also referred to as accessory cells, to generate a cell cluster that is not only able to accurately secrete insulin in response to glucose, but also superior in terms of other key features (e.g. maintaining a vasculature, longer durability in vivo and not necessitating immunosuppression after transplantation). Over the past decade, numerous 3D cell culture techniques have been integrated to create an engineered heterocellular islet organoid that addresses current obstacles. Here, we first discuss the different cell types used to prepare heterocellular organoids for islet transplantation and their contribution to the organoids design. We then introduce various cell culture techniques that are incorporated to prepare a fully functional and insulin secreting organoids with select features. Finally, we discuss the challenges and present a future outlook for improving clinical outcomes of islet transplantation.
Collapse
Affiliation(s)
- M Birgul Akolpoglu
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Yasemin Inceoglu
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Ugur Bozuyuk
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey
| | - Ana Rita Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials. University of Aveiro. Campus Universitário de Santiago. 3810-193 Aveiro. Portugal
| | - Seda Kizilel
- Chemical and Biological Engineering, Koc University, Sariyer, 34450, Istanbul, Turkey.
| |
Collapse
|
11
|
Lin EE, Scott-Solomon E, Kuruvilla R. Peripheral Innervation in the Regulation of Glucose Homeostasis. Trends Neurosci 2020; 44:189-202. [PMID: 33229051 DOI: 10.1016/j.tins.2020.10.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/07/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Precise regulation of circulating glucose is crucial for human health and ensures a sufficient supply to the brain, which relies almost exclusively on glucose for metabolic energy. Glucose homeostasis is coordinated by hormone-secreting endocrine cells in the pancreas, as well as glucose utilization and production in peripheral metabolic tissues including the liver, muscle, and adipose tissue. Glucose-regulatory tissues receive dense innervation from sympathetic, parasympathetic, and sensory fibers. In this review, we summarize the functions of peripheral nerves in glucose regulation and metabolism. Dynamic changes in peripheral innervation have also been observed in animal models of obesity and diabetes. Together, these studies highlight the importance of peripheral nerves as a new therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Eugene E Lin
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
12
|
Alkan I, Altunkaynak BZ, Kivrak EG, Kaplan AA, Arslan G. Is vagal stimulation or inhibition benefit on the regulation of the stomach brain axis in obesity? Nutr Neurosci 2020; 25:758-770. [PMID: 33034260 DOI: 10.1080/1028415x.2020.1809875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Objective: Possible effects of the vagus inhibition and stimulation on the hypothalamic nuclei, myenteric plexes and the vagus nerve were investigated.Methods: The female rats divided to the inhibition (INH), stimulation (STI) and, sham (SHAM) groups were fed with high fat diet (including 40% of energy from animal fat). After nine weeks, the rats were allowed to recover for 4 weeks in INH group. In STI group, the left vagus nerve stimulated (30 Hz/500 msn/30 sec.) starting 2nd post operative day for 5 minutes during 4 weeks. Healthy female rats used as control (CONT). Then, tissue samples were analyzed by biochemical, histological and stereological methods.Results: The mean number of the neurons in the arcuate nucleus of the INH group was significantly less; but, that is significantly more in the STI group compared to the other groups. The neuronal density of ventromedial nucleus in the STI group was higher; while the density in the INH group was lower than the other groups. In the dorsomedial nucleus, neuron density of the INH group was lower than the other groups. In terms of the myenteric plexus volumes, that of the INH group was lowest. The myelinated axon number in the INH group was significantly highest. The myelin sheath thickness and axon area of the INH group was significantly lower than the other groups.Discussion: The results of the study show that the vagal inhibition is more effective than the vagal stimulation on the weight loss in the obesity.
Collapse
Affiliation(s)
- Işınsu Alkan
- Department of Histology and Embryology, Faculty of Medicine, İstanbul Okan University, İstanbul, Turkey
| | - Berrin Zuhal Altunkaynak
- Department of Histology and Embryology, Faculty of Medicine, İstanbul Okan University, İstanbul, Turkey
| | - Elfide Gizem Kivrak
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| | - Arife Ahsen Kaplan
- Department of Histology and Embryology, Faculty of Medicine, Medipol University, Istanbul, Turkey
| | - Gülay Arslan
- Department of Histology and Embryology, Faculty of Medicine, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
13
|
Abstract
The islet of Langerhans is a complex endocrine micro-organ consisting of a multitude of endocrine and non-endocrine cell types. The two most abundant and prominent endocrine cell types, the beta and the alpha cells, are essential for the maintenance of blood glucose homeostasis. While the beta cell produces insulin, the only blood glucose-lowering hormone of the body, the alpha cell releases glucagon, which elevates blood glucose. Under physiological conditions, these two cell types affect each other in a paracrine manner. While the release products of the beta cell inhibit alpha cell function, the alpha cell releases factors that are stimulatory for beta cell function and increase glucose-stimulated insulin secretion. The aim of this review is to provide a comprehensive overview of recent research into the regulation of beta cell function by alpha cells, focusing on the effect of alpha cell-secreted factors, such as glucagon and acetylcholine. The consequences of differences in islet architecture between species on the interplay between alpha and beta cells is also discussed. Finally, we give a perspective on the possibility of using an in vivo imaging approach to study the interactions between human alpha and beta cells under in vivo conditions. Graphical abstract.
Collapse
Affiliation(s)
- Tilo Moede
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska Sjukhuset L1:03, 17176, Stockholm, Sweden.
| | - Ingo B Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska Sjukhuset L1:03, 17176, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska Sjukhuset L1:03, 17176, Stockholm, Sweden
| |
Collapse
|
14
|
Payne SC, Ward G, MacIsaac RJ, Hyakumura T, Fallon JB, Villalobos J. Differential effects of vagus nerve stimulation strategies on glycemia and pancreatic secretions. Physiol Rep 2020; 8:e14479. [PMID: 32512650 PMCID: PMC7280012 DOI: 10.14814/phy2.14479] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 12/14/2022] Open
Abstract
Despite advancements in pharmacotherapies, glycemia is poorly controlled in type 2 diabetic patients. As the vagus nerve regulates energy metabolism, here we evaluated the effect various electrical vagus nerve stimulation strategies have on glycemia and glucose-regulating hormones, as a first step to developing a novel therapy of type 2 diabetes. Sprague-Dawley rats were anesthetized, the abdominal (anterior) vagus nerve implanted, and various stimulation strategies applied to the nerve: (a) 15 Hz; (b) 4 kHz, or 40 kHz and; (c) a combination of 15 Hz and 40 kHz to directionally activate afferent or efferent vagal fibers. Following a glucose bolus (500 mg/kg, I.V.), stimulation strategies were applied (60 min) and serial blood samples taken. No stimulation was used as a crossover control sequence. Applying 15 Hz stimulation significantly increased glucose (+2.9 ± 0.2 mM·hr, p = .015) and glucagon (+17.1 ± 8.0 pg·hr/ml, p = .022), compared to no stimulation. Application of 4 kHz stimulation also significantly increased glucose levels (+1.5 ± 0.5 mM·hr, p = .049), while 40 kHz frequency stimulation resulted in no changes to glucose levels but did significantly lower glucagon (-12.3 ± 1.1 pg·hr/ml, p = .0009). Directional afferent stimulation increased glucose (+2.4 ± 1.5 mM·hr) and glucagon levels (+39.5 ± 15.0 pg·hr/ml). Despite hyperglycemia resulting when VNS, aVNS, and 4 kHz stimulation strategies were applied, the changes in insulin levels were not significant (p ≥ .05). In summary, vagus nerve stimulation modulates glycemia by effecting glucagon and insulin secretions, and high-frequency 40 kHz stimulation may have potential application for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Sophie C. Payne
- Bionics InstituteEast MelbourneVic.Australia
- Medical Bionics DepartmentThe University of MelbourneParkvilleVic.Australia
| | - Glenn Ward
- Bionics InstituteEast MelbourneVic.Australia
- Department of Endocrinology and DiabetesSt Vincent’s HospitalFitzroyVic.Australia
- Department of MedicineThe University of MelbourneParkvilleVic.Australia
| | - Richard J. MacIsaac
- Bionics InstituteEast MelbourneVic.Australia
- Department of Endocrinology and DiabetesSt Vincent’s HospitalFitzroyVic.Australia
- Department of MedicineThe University of MelbourneParkvilleVic.Australia
| | - Tomoko Hyakumura
- Bionics InstituteEast MelbourneVic.Australia
- Medical Bionics DepartmentThe University of MelbourneParkvilleVic.Australia
| | - James B. Fallon
- Bionics InstituteEast MelbourneVic.Australia
- Medical Bionics DepartmentThe University of MelbourneParkvilleVic.Australia
| | - Joel Villalobos
- Bionics InstituteEast MelbourneVic.Australia
- Medical Bionics DepartmentThe University of MelbourneParkvilleVic.Australia
| |
Collapse
|
15
|
Rodriguez-Diaz R, Tamayo A, Hara M, Caicedo A. The Local Paracrine Actions of the Pancreatic α-Cell. Diabetes 2020; 69:550-558. [PMID: 31882565 PMCID: PMC7085245 DOI: 10.2337/dbi19-0002] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Secretion of glucagon from the pancreatic α-cells is conventionally seen as the first and most important defense against hypoglycemia. Recent findings, however, show that α-cell signals stimulate insulin secretion from the neighboring β-cell. This article focuses on these seemingly counterintuitive local actions of α-cells and describes how they impact islet biology and glucose metabolism. It is mostly based on studies published in the last decade on the physiology of α-cells in human islets and incorporates results from rodents where appropriate. As this and the accompanying articles show, the emerging picture of α-cell function is one of increased complexity that needs to be considered when developing new therapies aimed at promoting islet function in the context of diabetes.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Tamayo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, IL
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
16
|
Noguchi GM, Huising MO. Integrating the inputs that shape pancreatic islet hormone release. Nat Metab 2019; 1:1189-1201. [PMID: 32694675 PMCID: PMC7378277 DOI: 10.1038/s42255-019-0148-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
The pancreatic islet is a complex mini organ composed of a variety of endocrine cells and their support cells, which together tightly control blood glucose homeostasis. Changes in glucose concentration are commonly regarded as the chief signal controlling insulin-secreting beta cells, glucagon-secreting alpha cells and somatostatin-secreting delta cells. However, each of these cell types is highly responsive to a multitude of endocrine, paracrine, nutritional and neural inputs, which collectively shape the final endocrine output of the islet. Here, we review the principal inputs for each islet-cell type and the physiological circumstances in which these signals arise, through the prism of the insights generated by the transcriptomes of each of the major endocrine-cell types. A comprehensive integration of the factors that influence blood glucose homeostasis is essential to successfully improve therapeutic strategies for better diabetes management.
Collapse
Affiliation(s)
- Glyn M Noguchi
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA.
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
17
|
Vagus-macrophage-hepatocyte link promotes post-injury liver regeneration and whole-body survival through hepatic FoxM1 activation. Nat Commun 2018; 9:5300. [PMID: 30546054 PMCID: PMC6294142 DOI: 10.1038/s41467-018-07747-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022] Open
Abstract
The liver possesses a high regenerative capacity. Liver regeneration is a compensatory response overcoming disturbances of whole-body homeostasis provoked by organ defects. Here we show that a vagus-macrophage-hepatocyte link regulates acute liver regeneration after liver injury and that this system is critical for promoting survival. Hepatic Foxm1 is rapidly upregulated after partial hepatectomy (PHx). Hepatic branch vagotomy (HV) suppresses this upregulation and hepatocyte proliferation, thereby increasing mortality. In addition, hepatic FoxM1 supplementation in vagotomized mice reverses the suppression of liver regeneration and blocks the increase in post-PHx mortality. Hepatic macrophage depletion suppresses both post-PHx Foxm1 upregulation and remnant liver regeneration, and increases mortality. Hepatic Il-6 rises rapidly after PHx and this is suppressed by HV, muscarinic blockade or resident macrophage depletion. Furthermore, IL-6 neutralization suppresses post-PHx Foxm1 upregulation and remnant liver regeneration. Collectively, vagal signal-mediated IL-6 production in hepatic macrophages upregulates hepatocyte FoxM1, leading to liver regeneration and assures survival. The mechanisms underlying the regenerative capacity of the liver are not fully understood. Here, the authors show that the acute regenerative response to liver injury in mice is regulated by the communication involving the vagus nerve, macrophages, and hepatocytes, leading to hepatic FoxM1 activation and promotion of overall survival.
Collapse
|
18
|
Bou Karam J, Cai W, Mohamed R, Huang T, Meng L, Homan EP, Dirice E, Kahn CR, El Ouaamari A. TRPV1 neurons regulate β-cell function in a sex-dependent manner. Mol Metab 2018; 18:60-67. [PMID: 30473098 PMCID: PMC6308974 DOI: 10.1016/j.molmet.2018.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/15/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023] Open
Abstract
There is emerging evidence to support an important role for the transient receptor potential vanilloid type 1 (TRPV1) sensory innervation in glucose homeostasis. However, it remains unknown whether the glucoregulatory action of these afferent neurons is sex-biased and whether it is pancreatic β-cell-mediated. OBJECTIVE We investigated in male and female mice whether denervation of whole-body or pancreas-projecting TRPV1 sensory neurons regulates adult functional β-cell mass and alters systemic glucose homeostasis. METHODS We used a combination of pharmacological and surgical approaches to ablate whole-body or pancreatic TRPV1 sensory neurons and assessed islet β-cell function and mass, aspects of glucose and insulin homeostasis, and energy expenditure. RESULTS Capsaicin-induced chemodenervation of whole-body TRPV1 sensory neurons improved glucose clearance and enhanced glucose-stimulated insulin secretion without alterations in β-cell proliferation and mass, systemic insulin sensitivity, body composition, and energy expenditure. Similarly, denervation of intrapancreatic TRPV1 afferents by pancreas intraductal injection of capsaicin or surgical removal of the dorsal root ganglia projecting into the pancreas lowered post-absorptive glucose levels and increased insulin release upon glucose stimulation. The beneficial effects of TRPV1 sensory denervation on glucose tolerance and β-cell function were observed in male but not female mice. CONCLUSION Collectively, these findings suggest that TRPV1 neurons regulate glucose homeostasis, at least partly, through direct modulation of glucose-induced insulin secretion and that this regulation operates in a sex-dependent manner.
Collapse
Affiliation(s)
- Joey Bou Karam
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Weikang Cai
- Section in Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Rowaida Mohamed
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Tianwen Huang
- Dana-Farber Cancer Institute and Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Lingqiong Meng
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Erica Paige Homan
- Section in Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Ercument Dirice
- Section of Islet Cell and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, USA
| | - C Ronald Kahn
- Section in Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - Abdelfattah El Ouaamari
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Güemes A, Georgiou P. Review of the role of the nervous system in glucose homoeostasis and future perspectives towards the management of diabetes. Bioelectron Med 2018; 4:9. [PMID: 32232085 PMCID: PMC7098234 DOI: 10.1186/s42234-018-0009-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/10/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetes is a disease caused by a breakdown in the glucose metabolic process resulting in abnormal blood glucose fluctuations. Traditionally, control has involved external insulin injection in response to elevated blood glucose to substitute the role of the beta cells in the pancreas which would otherwise perform this function in a healthy individual. The central nervous system (CNS), however, also plays a vital role in glucose homoeostasis through the control of pancreatic secretion and insulin sensitivity which could potentially be used as a pathway for enhancing glucose control. In this review, we present an overview of the brain regions, peripheral nerves and molecular mechanisms by which the CNS regulates glucose metabolism and the potential benefits of modulating them for diabetes management. Development of technologies to interface to the nervous system will soon become a reality through bioelectronic medicine and we present the emerging opportunities for the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Amparo Güemes
- Centre for Bio-Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Pantelis Georgiou
- Centre for Bio-Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, South Kensington Campus, London, UK
| |
Collapse
|
20
|
Cholinergic signaling mediates the effects of xenin-25 on secretion of pancreatic polypeptide but not insulin or glucagon in humans with impaired glucose tolerance. PLoS One 2018; 13:e0192441. [PMID: 29466430 PMCID: PMC5821323 DOI: 10.1371/journal.pone.0192441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 01/10/2018] [Indexed: 01/14/2023] Open
Abstract
We previously demonstrated that infusion of an intestinal peptide called xenin-25 (Xen) amplifies the effects of glucose-dependent insulinotropic polypeptide (GIP) on insulin secretion rates (ISRs) and plasma glucagon levels in humans. However, these effects of Xen, but not GIP, were blunted in humans with type 2 diabetes. Thus, Xen rather than GIP signaling to islets fails early during development of type 2 diabetes. The current crossover study determines if cholinergic signaling relays the effects of Xen on insulin and glucagon release in humans as in mice. Fasted subjects with impaired glucose tolerance were studied. On eight separate occasions, each person underwent a single graded glucose infusion- two each with infusion of albumin, Xen, GIP, and GIP plus Xen. Each infusate was administered ± atropine. Heart rate and plasma glucose, insulin, C-peptide, glucagon, and pancreatic polypeptide (PP) levels were measured. ISRs were calculated from C-peptide levels. All peptides profoundly increased PP responses. From 0 to 40 min, peptide(s) infusions had little effect on plasma glucose concentrations. However, GIP, but not Xen, rapidly and transiently increased ISRs and glucagon levels. Both responses were further amplified when Xen was co-administered with GIP. From 40 to 240 min, glucose levels and ISRs continually increased while glucagon concentrations declined, regardless of infusate. Atropine increased resting heart rate and blocked all PP responses but did not affect ISRs or plasma glucagon levels during any of the peptide infusions. Thus, cholinergic signaling mediates the effects of Xen on insulin and glucagon release in mice but not humans.
Collapse
|
21
|
Abstract
BACKGROUND Recently, the US FDA has approved "vagal blocking therapy or vBLoc® therapy" as a new treatment for obesity. The aim of the present study was to study the mechanism-of-action of "VBLOC" in rat models. METHODS Rats were implanted with VBLOC, an intra-abdominal electrical device with leads placed around gastric vagal trunks through an abdominal incision and controlled by wireless device. Body weight, food intake, hunger/satiety, and metabolic parameters were monitored by a comprehensive laboratory animal monitoring system. Brain-gut responses were analyzed physiologically. RESULTS VBLOC reduced body weight and food intake, which was associated with increased satiety but not with decreased hunger. Brain activities in response to VBLOC included increased gene expression of leptin and CCKb receptors, interleukin-1β, tumor necrosis factor, and transforming growth factor β1 in the brainstem; increased CCK, somatostatin, and tyrosine hydroxylase in the hippocampus; increased NPY, AgRP, and Foxa2 in the hypothalamus; and reduced CCKb receptor, melanocortin 4 receptor, and insulin receptor in the hypothalamus. Plasma concentrations of CCK, gastrin, glucagon, GLP-1, and PYY and gastric acid secretion were unchanged in response to VBLOC. CONCLUSIONS Based on the present study, we may suggest that VBLOC induces satiety through vagal signaling, leading to reduced food intake and loss of body weight.
Collapse
|
22
|
Abstract
Type 1 diabetes (T1D) patients who receive pancreatic islet transplant experience significant improvement in their quality-of-life. This comes primarily through improved control of blood sugar levels, restored awareness of hypoglycemia, and prevention of serious and potentially life-threatening diabetes-associated complications, such as kidney failure, heart and vascular disease, stroke, nerve damage, and blindness. Therefore, beta cell replacement through transplantation of isolated islets is an important option in the treatment of T1D. However, lasting success of this promising therapy depends on durable survival and efficacy of the transplanted islets, which are directly influenced by the islet isolation procedures. Thus, isolating pancreatic islets with consistent and reliable quality is critical in the clinical application of islet transplantation.Quality of isolated islets is important in pre-clinical studies as well, as efforts to advance and improve clinical outcomes of islet transplant therapy have relied heavily on animal models ranging from rodents, to pigs, to nonhuman primates. As a result, pancreatic islets have been isolated from these and other species and used in a variety of in vitro or in vivo applications for this and other research purposes. Protocols for islet isolation have been somewhat similar across species, especially, in mammals. However, given the increasing evidence about the distinct structural and functional features of human and mouse islets, using similar methods of islet isolation may contribute to inconsistencies in the islet quality, immunogenicity, and experimental outcomes. This may also contribute to the discrepancies commonly observed between pre-clinical findings and clinical outcomes. Therefore, it is prudent to consider the particular features of pancreatic islets from different species when optimizing islet isolation protocols.In this chapter, we explore the structural and functional features of pancreatic islets from mice, pigs, nonhuman primates, and humans because of their prevalent use in nonclinical, preclinical, and clinical applications.
Collapse
|
23
|
Chowdhury S, Wang S, Dunai J, Kilpatrick R, Oestricker LZ, Wallendorf MJ, Patterson BW, Reeds DN, Wice BM. Hormonal Responses to Cholinergic Input Are Different in Humans with and without Type 2 Diabetes Mellitus. PLoS One 2016; 11:e0156852. [PMID: 27304975 PMCID: PMC4909255 DOI: 10.1371/journal.pone.0156852] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/20/2016] [Indexed: 12/14/2022] Open
Abstract
Peripheral muscarinic acetylcholine receptors regulate insulin and glucagon release in rodents but their importance for similar roles in humans is unclear. Bethanechol, an acetylcholine analogue that does not cross the blood-brain barrier, was used to examine the role of peripheral muscarinic signaling on glucose homeostasis in humans with normal glucose tolerance (NGT; n = 10), impaired glucose tolerance (IGT; n = 11), and type 2 diabetes mellitus (T2DM; n = 9). Subjects received four liquid meal tolerance tests, each with a different dose of oral bethanechol (0, 50, 100, or 150 mg) given 60 min before a meal containing acetaminophen. Plasma pancreatic polypeptide (PP), glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), glucose, glucagon, C-peptide, and acetaminophen concentrations were measured. Insulin secretion rates (ISRs) were calculated from C-peptide levels. Acetaminophen and PP concentrations were surrogate markers for gastric emptying and cholinergic input to islets. The 150 mg dose of bethanechol increased the PP response 2-fold only in the IGT group, amplified GLP-1 release in the IGT and T2DM groups, and augmented the GIP response only in the NGT group. However, bethanechol did not alter ISRs or plasma glucose, glucagon, or acetaminophen concentrations in any group. Prior studies showed infusion of xenin-25, an intestinal peptide, delays gastric emptying and reduces GLP-1 release but not ISRs when normalized to plasma glucose levels. Analysis of archived plasma samples from this study showed xenin-25 amplified postprandial PP responses ~4-fold in subjects with NGT, IGT, and T2DM. Thus, increasing postprandial cholinergic input to islets augments insulin secretion in mice but not humans. Trial Registration: ClinicalTrials.gov NCT01434901
Collapse
Affiliation(s)
- Sara Chowdhury
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Songyan Wang
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Judit Dunai
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Rachel Kilpatrick
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Lauren Z. Oestricker
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Michael J. Wallendorf
- Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Bruce W. Patterson
- Department of Internal Medicine, Division of Nutritional Science, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Dominic N. Reeds
- Department of Internal Medicine, Division of Nutritional Science, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
| | - Burton M. Wice
- Department of Internal Medicine, Division of Endocrinology, Metabolism and Lipid Research Washington University School of Medicine, Saint Louis, MO, United States of America
- * E-mail:
| |
Collapse
|
24
|
Galanin regulates blood glucose level in the zebrafish: a morphological and functional study. Histochem Cell Biol 2015; 145:105-17. [PMID: 26496922 PMCID: PMC4710661 DOI: 10.1007/s00418-015-1376-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2015] [Indexed: 02/02/2023]
Abstract
The present study has demonstrated the galaninergic innervation of the endocrine pancreas including sources of the galaninergic nerve fibers, and the influence of galanin receptor agonists on blood glucose level in the zebrafish. For the first time, a very abundant galaninergic innervation of the endocrine pancreas during development is shown, from the second day post-fertilization to adulthood. The fibers originated from ganglia consisting of galanin-IR, non-adrenergic (non-sensory) neurons located rostrally to the pancreatic tissue. The ganglia were found on the dorsal side of the initial part of the anterior intestinal segment, close to the intestinal branch of the vagus nerve. The galanin-IR neurons did not show immunoreactivity for applied antibodies against tyrosine hydroxylase, choline acetyltransferase, and vesicular acetylcholine transporter. Intraperitoneal injections of galanin analog NAX 5055 resulted in a statistically significant increase in the blood glucose level. Injections of another galanin receptor agonist, galnon, also caused a rise in blood glucose level; however, it was not statistically significant. The present findings suggest that, like in mammals, in the zebrafish galanin is involved in the regulation of blood glucose level. However, further studies are needed to elucidate the exact mechanism of the galanin action.
Collapse
|
25
|
Barreto SG. How does cigarette smoking cause acute pancreatitis? Pancreatology 2015; 16:157-63. [PMID: 26419886 DOI: 10.1016/j.pan.2015.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 08/22/2015] [Accepted: 09/03/2015] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Acute Pancreatitis (AP) is an emerging health problem world-wide and it is a major cause of admissions for gastrointestinal disease in many countries. Amongst the more common causes (alcohol and gallstones), recent evidence has emerged indicating that smoking is an independent risk factor for AP. However, the mechanisms involved in smoking-induced AP have not been completely elucidated. This review puts together all the published evidence in literature to present the clinical and laboratory evidence relating smoking to the causation of AP. DISCUSSION The two main metabolites from cigarette smoke, namely nicotine and NNK are able to induce functional and histological changes within the pancreas consistent with AP. The major mechanisms involved include their action on acinar cells and zymogen secretion through pathways involving CCK and the nicotinic preganglionic receptors. Effects on the pancreatic microvasculature may be mediated through the nitric oxide pathway. There is indirect evidence to suggest that nicotine and acrolein may lead to CFTR dysfunction thereby influencing ductal secretion. However, direct evidence for this effect is needed. The effect of cigarette smoke metabolites on stellate cells and the islets warrants further investigation in the context of pathogenesis of AP. CONCLUSION Using a step-wise approach, the review revisits the effects of the various metabolites of cigarette smoke on the constituents of the pancreas (exocrine, endocrine, neurohormonal, stellate cells, ductal system) and highlights their proven, and potential, mechanisms in triggering off an attack of AP.
Collapse
Affiliation(s)
- Savio G Barreto
- Department of Gastrointestinal Surgery, Gastrointestinal Oncology, and Bariatric Surgery, Medanta Institute of Digestive and Hepatobiliary Sciences, Medanta, The Medicity, Sector 38, Gurgaon, Haryana, India.
| |
Collapse
|
26
|
Dhumpa R, Truong TM, Wang X, Bertram R, Roper MG. Negative feedback synchronizes islets of Langerhans. Biophys J 2014; 106:2275-82. [PMID: 24853756 DOI: 10.1016/j.bpj.2014.04.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 04/01/2014] [Accepted: 04/11/2014] [Indexed: 01/19/2023] Open
Abstract
Insulin is released from the pancreas in pulses with a period of ~ 5 min. These oscillatory insulin levels are essential for proper liver utilization and perturbed pulsatility is observed in type 2 diabetes. What coordinates the many islets of Langerhans throughout the pancreas to produce unified oscillations of insulin secretion? One hypothesis is that coordination is achieved through an insulin-dependent negative feedback action of the liver onto the glucose level. This hypothesis was tested in an in vitro setting using a microfluidic system where the population response from a group of islets was input to a model of hepatic glucose uptake, which provided a negative feedback to the glucose level. This modified glucose level was then delivered back to the islet chamber where the population response was again monitored and used to update the glucose concentration delivered to the islets. We found that, with appropriate parameters for the model, oscillations in islet activity were synchronized. This approach demonstrates that rhythmic activity of a population of physically uncoupled islets can be coordinated by a downstream system that senses islet activity and supplies negative feedback. In the intact animal, the liver can play this role of the coordinator of islet activity.
Collapse
Affiliation(s)
- Raghuram Dhumpa
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306
| | - Tuan M Truong
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306
| | - Xue Wang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306
| | - Richard Bertram
- Department of Mathematics and Program in Neuroscience, Florida State University, Tallahassee, FL 32306; Program in Molecular Biophysics, Florida State University, Tallahassee, FL 32306.
| | - Michael G Roper
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306; Program in Molecular Biophysics, Florida State University, Tallahassee, FL 32306.
| |
Collapse
|
27
|
Abstract
The autonomic nervous system affects glucose metabolism partly through its connection to the pancreatic islet. Since its discovery by Paul Langerhans, the precise innervation patterns of the islet has remained elusive, mainly because of technical limitations. Using 3-dimensional reconstructions of axonal terminal fields, recent studies have determined the innervation patterns of mouse and human islets. In contrast to the mouse islet, endocrine cells within the human islet are sparsely contacted by autonomic axons. Instead, the invading sympathetic axons preferentially innervate smooth muscle cells of blood vessels. This innervation pattern suggests that, rather than acting directly on endocrine cells, sympathetic nerves may control hormone secretion by modulating blood flow in human islets. In addition to autonomic efferent axons, islets also receive sensory innervation. These axons transmit sensory information to the brain but also have the ability to locally release neuroactive substances that have been suggested to promote diabetes pathogenesis. We discuss recent findings on islet innervation, the connections of the islet with the brain, and the role islet innervation plays during the progression of diabetes.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Rolf Luft Research Center for Diabetes & Endocrinology, Karolinska Institutet, Stockholm, SE-17177, Sweden; Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Alejandro Caicedo
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
28
|
Tang SC, Peng SJ, Chien HJ. Imaging of the islet neural network. Diabetes Obes Metab 2014; 16 Suppl 1:77-86. [PMID: 25200300 DOI: 10.1111/dom.12342] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/28/2014] [Indexed: 02/06/2023]
Abstract
The islets of Langerhans receive signals from the circulation and nerves to modulate hormone secretion in response to physiological cues. Although the rich islet innervation has been documented in the literature dating as far back as Paul Langerhans' discovery of islets in the pancreas, it remains a challenging task for researchers to acquire detailed islet innervation patterns in health and disease due to the dispersed nature of the islet neurovascular network. In this article, we discuss the recent development of 3-dimensional (3D) islet neurohistology, in which transparent pancreatic specimens were prepared by optical clearing to visualize the islet microstructure, vasculature and innervation with deep-tissue microscopy. Mouse islets were used as an example to illustrate how to apply this 3D imaging approach to characterize (i) the islet parasympathetic innervation, (ii) the islet sympathetic innervation and its reinnervation after transplantation under the kidney capsule and (iii) the reactive cellular response of the Schwann cell network in islet injury. While presenting and characterizing the innervation patterns, we also discuss how to apply the signals derived from transmitted light microscopy, vessel painting and immunostaining of neural markers to verify the location and source of tissue information. In summary, the systematic development of tissue labelling, clearing and imaging methods to reveal the islet neuroanatomy offers insights to help study the neural-islet regulatory mechanisms and the role of neural tissue remodelling in the development of diabetes.
Collapse
Affiliation(s)
- S-C Tang
- Connectomics Research Center, National Tsing Hua University, Hsinchu, Taiwan; Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | | | | |
Collapse
|
29
|
Lin CL, Williams L, Seki Y, Kaur H, Hartil K, Fiallo A, Glenn AS, Katz EB, Charron MJ, Vuguin PM. Effects of genetics and in utero diet on murine pancreatic development. J Endocrinol 2014; 222:217-27. [PMID: 24895417 PMCID: PMC4287255 DOI: 10.1530/joe-14-0114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Intrauterine (IU) malnutrition could alter pancreatic development. In this study, we describe the effects of high-fat diet (HFD) during pregnancy on fetal growth and pancreatic morphology in an 'at risk' animal model of metabolic disease, the glucose transporter 4 (GLUT4) heterozygous mouse (G4+/-). WT female mice mated with G4+/- males were fed HFD or control diet (CD) for 2 weeks before mating and throughout pregnancy. At embryonic day 18.5, fetuses were killed and pancreata isolated for analysis of morphology and expression of genes involved in insulin (INS) cell development, proliferation, apoptosis, glucose transport and function. Compared with WT CD, WT HFD fetal pancreata had a 2.4-fold increase in the number of glucagon (GLU) cells (P=0.023). HFD also increased GLU cell size by 18% in WT pancreata compared with WT CD. Compared with WT CD, G4+/- CD had an increased number of INS cells and decreased INS and GLU cell size. Compared with G4+/- CD, G4+/- HFD fetuses had increased pancreatic gene expression of Igf2, a mitogen and inhibitor of apoptosis. The expression of genes involved in proliferation, apoptosis, glucose transport, and INS secretion was not altered in WT HFD compared with G4+/- HFD pancreata. In contrast to WT HFD pancreata, HFD exposure did not alter pancreatic islet morphology in fetuses with GLUT4 haploinsufficiency; this may be mediated in part by increased Igf2 expression. Thus, interactions between IU diet and fetal genetics may play a critical role in the developmental origins of health and disease.
Collapse
Affiliation(s)
- Chia-Lei Lin
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Lyda Williams
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Yoshinori Seki
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Harpreet Kaur
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USADepartments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USADepartments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Kirsten Hartil
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Ariana Fiallo
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - A Scott Glenn
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Ellen B Katz
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Maureen J Charron
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USADepartments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USADepartments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| | - Patricia M Vuguin
- Departments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USADepartments of PediatricsNeonatologyBiochemistryObstetrics and Gynecology and Women's HealthMedicineAlbert Einstein College of Medicine, 1300 Morris Park Avenue, F312, Bronx, New York 10461, USADepartment of PediatricsHofstra School of Medicine, Cohen Children's Medical Center, 1991 Marcus Avenue, Lake Success, New York 11402, USA
| |
Collapse
|
30
|
Yin J, Kuang J, Chandalia M, Tuvdendorj D, Tumurbaatar B, Abate N, Chen JDZ. Hypoglycemic effects and mechanisms of electroacupuncture on insulin resistance. Am J Physiol Regul Integr Comp Physiol 2014; 307:R332-9. [PMID: 24848362 DOI: 10.1152/ajpregu.00465.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to investigate effects and mechanisms of electroacupuncture (EA) on blood glucose and insulin sensitivity in mice fed a high-fat diet. Both wild-type (WT) and adipose ectonucleotide pyrophosphate phosphodiesterase (ENPP1) transgenic (TG) mice were fed a high-fat diet for 12 wk; for each mouse, an intraperitoneal glucose tolerance test (IPGTT) and insulin tolerance test (ITT) were performed with or without EA at abdomen or auricular areas. A high-fat diet-induced insulin resistance in both WT and TG mice. In the WT mice, EA at 3 Hz and 15 Hz, but not at 1 Hz or 100 Hz, via CV4+CV12 significantly reduced postprandial glucose levels; EA at 3 Hz was most potent. The glucose level was reduced by 61.7% at 60 min and 74.5% at 120 min with EA at 3 Hz (all P < 0.001 vs. control). Similar hypoglycemic effect was noted in the TG mice. On the contrary, EA at auricular points increased postprandial glucose level (P < 0.03). 4). EA at 3 Hz via CV4+CV12 significantly enhanced the decrease of blood glucose after insulin injection, suggesting improvement of insulin sensitivity. Plasma free fatty acid was significantly suppressed by 42.5% at 15 min and 50.8% at 30 min with EA (P < 0.01) in both WT and TG mice. EA improves glucose tolerance in both WT and TG mice fed a high-fat diet, and the effect is associated with stimulation parameters and acupoints and is probably attributed to the reduction of free fatty acid.
Collapse
Affiliation(s)
- Jieyun Yin
- Division of Gastroenterology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas
| | - Jian Kuang
- Division of Gastroenterology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas; Division of Endocrinology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China; and
| | - Manisha Chandalia
- Division of Endocrinology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas
| | - Demidmaa Tuvdendorj
- Division of Endocrinology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas
| | - Batbayar Tumurbaatar
- Division of Endocrinology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas
| | - Nicola Abate
- Division of Endocrinology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas
| | - Jiande D Z Chen
- Division of Gastroenterology, Department of Internal Medicine. University of Texas Medical Branch, Galveston, Texas; Ningbo Pace Translational Medical Research Center, Ningbo, China
| |
Collapse
|
31
|
Volpato AM, Zugno AI, Quevedo J. Recent evidence and potential mechanisms underlying weight gain and insulin resistance due to atypical antipsychotics. BRAZILIAN JOURNAL OF PSYCHIATRY 2014; 35:295-304. [PMID: 24142093 DOI: 10.1590/1516-4446-2012-1052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 12/20/2012] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Atypical antipsychotics (AAPs) promote obesity and insulin resistance. In this regard, the main objective of this study was to present potential mechanisms and evidence concerning side effects of atypical antipsychotics in humans and rodents. METHOD A systematic review of the literature was performed using the MEDLINE database. We checked the references of selected articles, review articles, and books on the subject. RESULTS This review provides consistent results concerning the side effects of olanzapine (OL) and clozapine (CLZ), whereas we found conflicting results related to other AAPs. Most studies involving humans describe the effects on body weight, adiposity, lipid profile, and blood glucose levels. However, it seems difficult to identify an animal model replicating the wide range of changes observed in humans. Animal lineage, route of administration, dose, and duration of treatment should be carefully chosen for the replication of the findings in humans. CONCLUSIONS Patients undergoing treatment with AAPs are at higher risk of developing adverse metabolic changes. This increased risk must be taken into account when making decisions about treatment. The influence of AAPs on multiple systems is certainly the cause of such effects. Specifically, muscarinic and histaminergic pathways seem to play important roles.
Collapse
Affiliation(s)
- Ana Maria Volpato
- Universidade do Extremo Sul Catarinense, Laboratory of Neurosciences, CriciúmaSC, Brazil
| | | | | |
Collapse
|
32
|
Muñoz-Bravo JL, Hidalgo-Figueroa M, Pascual A, López-Barneo J, Leal-Cerro A, Cano DA. GDNF is required for neural colonization of the pancreas. Development 2013; 140:3669-79. [PMID: 23903190 DOI: 10.1242/dev.091256] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The mammalian pancreas is densely innervated by both the sympathetic and parasympathetic nervous systems, which control exocrine and endocrine secretion. During embryonic development, neural crest cells migrating in a rostrocaudal direction populate the gut, giving rise to neural progenitor cells. Recent studies in mice have shown that neural crest cells enter the pancreatic epithelium at E11.5. However, the cues that guide the migration of neural progenitors into the pancreas are poorly defined. In this study we identify glial cell line-derived neurotrophic factor (GDNF) as a key player in this process. GDNF displays a dynamic expression pattern during embryonic development that parallels the chronology of migration and differentiation of neural crest derivatives in the pancreas. Conditional inactivation of Gdnf in the pancreatic epithelium results in a dramatic loss of neuronal and glial cells and in reduced parasympathetic innervation in the pancreas. Importantly, the innervation of other regions of the gut remains unaffected. Analysis of Gdnf mutant mouse embryos and ex vivo experiments indicate that GDNF produced in the pancreas acts as a neurotrophic factor for gut-resident neural progenitor cells. Our data further show that exogenous GDNF promotes the proliferation of pancreatic progenitor cells in organ culture. In summary, our results point to GDNF as crucial for the development of the intrinsic innervation of the pancreas.
Collapse
Affiliation(s)
- José Luis Muñoz-Bravo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Seville, Spain
| | | | | | | | | | | |
Collapse
|
33
|
Bonal CB, Baronnier DE, Pot C, Benkhoucha M, Schwab ME, Lalive PH, Herrera PL. Nogo-A downregulation improves insulin secretion in mice. Diabetes 2013; 62:1443-52. [PMID: 23274909 PMCID: PMC3636604 DOI: 10.2337/db12-0949] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is characterized by β-cell dysfunction and the subsequent depletion of insulin production, usually in a context of increased peripheral insulin resistance. T2D patients are routinely treated with oral antidiabetic agents such as sulfonylureas or dipeptidyl peptidase-4 antagonists, which promote glucose- and incretin-dependent insulin secretion, respectively. Interestingly, insulin secretion may also be induced by neural stimulation. Here we report the expression of Nogo-A in β-cells. Nogo-A is a membrane protein that inhibits neurite outgrowth and cell migration in the central nervous system. We observed that Nogo-A-deficient mice display improved insulin secretion and glucose clearance. This was associated with a stronger parasympathetic input and higher sensitivity of β-cells to the cholinergic analog carbachol. Insulin secretion was also improved in diabetic db/db mice treated with neutralizing antibody against Nogo-A. Together, these findings suggest that promoting the vagal stimulation of insulin secretion through the selective inhibition of Nogo-A could be a novel therapeutic approach in T2D.
Collapse
Affiliation(s)
- Claire B. Bonal
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Delphine E. Baronnier
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Caroline Pot
- Division of Neurology, Department of Neurosciences, Geneva University Hospital, Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Mahdia Benkhoucha
- Division of Neurology, Department of Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich, Zurich, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Patrice H. Lalive
- Division of Neurology, Department of Neurosciences, Geneva University Hospital, Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Department of Genetic and Laboratory Medicine, Geneva University Hospital, Geneva, Switzerland
| | - Pedro L. Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Corresponding author: Pedro L. Herrera,
| |
Collapse
|
34
|
Rodriguez-Diaz R, Caicedo A. Novel approaches to studying the role of innervation in the biology of pancreatic islets. Endocrinol Metab Clin North Am 2013; 42:39-56. [PMID: 23391238 PMCID: PMC3576136 DOI: 10.1016/j.ecl.2012.11.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The autonomic nervous system helps regulate glucose homeostasis by acting on pancreatic islets of Langerhans. Despite decades of research on the innervation of the pancreatic islet, the mechanisms used by the autonomic nervous input to influence islet cell biology have not been elucidated. This article discusses how these barriers can be overcome to study the role of the autonomic innervation of the pancreatic islet in glucose metabolism. It describes recent advances in microscopy and novel approaches to studying the effects of nervous input that may help clarify how autonomic axons regulate islet biology.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Diabetes Research Institute, Miller School of Medicine, University of Miami, FL 33136
| | - Alejandro Caicedo
- Diabetes Research Institute, Miller School of Medicine, University of Miami, FL 33136
- Department of Medicine, Miller School of Medicine, University of Miami, FL 33136
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, FL 33136
- Program in Neuroscience, Miller School of Medicine, University of Miami, FL 33136
| |
Collapse
|
35
|
Noninvasive in vivo model demonstrating the effects of autonomic innervation on pancreatic islet function. Proc Natl Acad Sci U S A 2012; 109:21456-61. [PMID: 23236142 DOI: 10.1073/pnas.1211659110] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The autonomic nervous system is thought to modulate blood glucose homeostasis by regulating endocrine cell activity in the pancreatic islets of Langerhans. The role of islet innervation, however, has remained elusive because the direct effects of autonomic nervous input on islet cell physiology cannot be studied in the pancreas. Here, we used an in vivo model to study the role of islet nervous input in glucose homeostasis. We transplanted islets into the anterior chamber of the eye and found that islet grafts became densely innervated by the rich parasympathetic and sympathetic nervous supply of the iris. Parasympathetic innervation was imaged intravitally by using transgenic mice expressing GFP in cholinergic axons. To manipulate selectively the islet nervous input, we increased the ambient illumination to increase the parasympathetic input to the islet grafts via the pupillary light reflex. This reduced fasting glycemia and improved glucose tolerance. These effects could be blocked by topical application of the muscarinic antagonist atropine to the eye, indicating that local cholinergic innervation had a direct effect on islet function in vivo. By using this approach, we found that parasympathetic innervation influences islet function in C57BL/6 mice but not in 129X1 mice, which reflected differences in innervation densities and may explain major strain differences in glucose homeostasis. This study directly demonstrates that autonomic axons innervating the islet modulate glucose homeostasis.
Collapse
|
36
|
Chiu YC, Hua TE, Fu YY, Pasricha PJ, Tang SC. 3-D imaging and illustration of the perfusive mouse islet sympathetic innervation and its remodelling in injury. Diabetologia 2012; 55:3252-61. [PMID: 22930160 DOI: 10.1007/s00125-012-2699-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 07/26/2012] [Indexed: 01/17/2023]
Abstract
AIMS/HYPOTHESIS Sympathetic nerves influence islet hormone levels in the circulation. Insights into islet sympathetic innervation and its remodelling in diabetes may impact future therapeutics. However, standard immunohistochemistry and microtome-based microscopy cannot provide an integral view of the islet neurovascular complex. We prepared transparent islet specimens to investigate the spatial relationship between sympathetic nerves, blood vessels and islet cells in normal, streptozotocin-injected and non-obese diabetic mouse models. METHODS Cardiac perfusion of fluorescent lectin was used to label pancreatic blood vessels. Tyrosine hydroxylase and nuclear staining were used to reveal islet sympathetic innervation and microstructure. Optical clearing (i.e. use of immersion solution to reduce scattering) was applied to enable 3-dimensional confocal microscopy of islets to visualise the sympathetic neurovascular complex in space. RESULTS Unlike previously reported morphology, we observed perfusive intra-islet, perivascular sympathetic innervation, in addition to peri-islet contacts of sympathetic nerves with alpha cells and sympathetic fibres encircling the adjacent arterioles. The intra-islet axons became markedly prominent in streptozotocin-injected mice (2 weeks after injection). In non-obese diabetic mice, lymphocytic infiltration remodelled the peri-islet sympathetic axons in early insulitis. CONCLUSIONS/INTERPRETATION We have established an imaging approach to reveal the spatial features of mouse islet sympathetic innervation. The neurovascular complex and sympathetic nerve-alpha cell contact suggest that sympathetic nerves modulate islet hormone secretion through blood vessels, in addition to acting directly on alpha cells. In islet injuries, sympathetic nerves undergo different remodelling in response to different pathophysiological cues.
Collapse
Affiliation(s)
- Y-C Chiu
- Connectomics Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | | | | | | | | |
Collapse
|
37
|
Simpson N, Maffei A, Freeby M, Burroughs S, Freyberg Z, Javitch J, Leibel RL, Harris PE. Dopamine-mediated autocrine inhibitory circuit regulating human insulin secretion in vitro. Mol Endocrinol 2012; 26:1757-72. [PMID: 22915827 DOI: 10.1210/me.2012-1101] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We describe a negative feedback autocrine regulatory circuit for glucose-stimulated insulin secretion in purified human islets in vitro. Using chronoamperometry and in vitro glucose-stimulated insulin secretion measurements, evidence is provided that dopamine (DA), which is loaded into insulin-containing secretory granules by vesicular monoamine transporter type 2 in human β-cells, is released in response to glucose stimulation. DA then acts as a negative regulator of insulin secretion via its action on D2R, which are also expressed on β-cells. We found that antagonism of receptors participating in islet DA signaling generally drive increased glucose-stimulated insulin secretion. These in vitro observations may represent correlates of the in vivo metabolic changes associated with the use of atypical antipsychotics, such as increased adiposity.
Collapse
Affiliation(s)
- Norman Simpson
- Division of Endocrinology, Department of Medicine, Columbia University Medical College, 650 West 168th Street, BB 2006, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Hammon HM, Steinhoff-Wagner J, Schönhusen U, Metges CC, Blum JW. Energy metabolism in the newborn farm animal with emphasis on the calf: endocrine changes and responses to milk-born and systemic hormones. Domest Anim Endocrinol 2012; 43:171-85. [PMID: 22480719 DOI: 10.1016/j.domaniend.2012.02.005] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/23/2012] [Accepted: 02/27/2012] [Indexed: 01/10/2023]
Abstract
Neonatal mammals need adaption to changes in nutrient supply because energy intake shifts from continuous parenteral supply of nutrients (mainly glucose, lactate, and amino acids) via the placenta to discontinuous colostrum and milk intake with lactose and fat as main energy sources. Besides ingested lactose, endogenous glucose production is essential in the neonate to assure sufficient glucose availability. Fetal endogenous glucose production is low, but endocrine changes (especially the prenatal rise of glucocorticoid production) promote maturation of metabolic pathways that enable marked glycogen synthesis before and enhanced gluconeogenesis after birth to establish an adequate glucose status during postnatal maturation. In preterm born farm animals gluconeogenic activity is low, mainly because of a low glucocorticoid and thyroid status. In full-term neonates, endogenous glucose production increases with age. Colostral bioactive components (such as growth factors, hormones, bioactive peptides, and cytokines) do not have a direct effect on endogenous glucose production. However, colostrum feeding stimulates intestinal growth and development, an effect at least in part mediated by bioactive substances. Increased nutrient and glucose absorption thus allows increased glucose supply and hepatic glycogen storage, which improves the glucose status. The improved energetic status of colostrum-fed neonates is reflected by an accelerated maturation of the somatotropic axis, leading especially to enhanced production of IGF-I in the neonate. Secretion and production of hormones involved in the regulation of glucose and fat metabolism in neonates depend on the developmental stage and the response to feeding. In addition, many such hormones have actions in the neonate that differ from adult animals. Endocrine action to support endogenous energy supply in neonates is probably not fully established, and therefore, needs postnatal maturation. Therefore, our knowledge on energy metabolism in the neonate needs to be extended to better understand the function and the failure and to assess endocrine responses during the neonatal period.
Collapse
Affiliation(s)
- H M Hammon
- Leibniz Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany.
| | | | | | | | | |
Collapse
|
39
|
Grouwels G, Vasylovska S, Olerud J, Leuckx G, Ngamjariyawat A, Yuchi Y, Jansson L, Van de Casteele M, Kozlova EN, Heimberg H. Differentiating neural crest stem cells induce proliferation of cultured rodent islet beta cells. Diabetologia 2012; 55:2016-25. [PMID: 22618811 DOI: 10.1007/s00125-012-2542-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 02/22/2012] [Indexed: 12/20/2022]
Abstract
AIMS/HYPOTHESIS Efficient stimulation of cycling activity in cultured beta cells would allow the design of new strategies for cell therapy in diabetes. Neural crest stem cells (NCSCs) play a role in beta cell development and maturation and increase the beta cell number in co-transplants. The mechanism behind NCSC-induced beta cell proliferation and the functional capacity of the new beta cells is not known. METHODS We developed a new in vitro co-culture system that enables the dissection of the elements that control the cellular interactions that lead to NCSC-dependent increase in islet beta cells. RESULTS Mouse NCSCs were cultured in vitro, first in medium that stimulated their proliferation, then under conditions that supported their differentiation. When mouse islet cells were cultured together with the NCSCs, more than 35% of the beta cells showed cycle activity. This labelling index is more than tenfold higher than control islets cultured without NCSCs. Beta cells that proliferated under these culture conditions were fully glucose responsive in terms of insulin secretion. NCSCs also induced beta cell proliferation in islets isolated from 1-year-old mice, but not in dissociated islet cells isolated from human donor pancreas tissue. To stimulate beta cell proliferation, NCSCs need to be in intimate contact with the beta cells. CONCLUSIONS/INTERPRETATION Culture of islet cells in contact with NCSCs induces highly efficient beta cell proliferation. The reported culture system is an excellent platform for further dissection of the minimal set of factors needed to drive this process and explore its potential for translation to diabetes therapy.
Collapse
Affiliation(s)
- G Grouwels
- Diabetes Research Center, Vrije Universiteit Brussel, Laarbeeklaan 103, B1090 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The pancreas is characterized by a major component, an exocrine and ductal system involved in digestion, and a minor component, the endocrine islets represented by islet micro-organs that tightly regulate glucose homoeostasis. Pancreatic organogenesis is strictly co-ordinated by transcription factors that are expressed sequentially to yield functional islets capable of maintaining glucose homoeostasis. Angiogenesis and innervation complete islet development, equipping islets to respond to metabolic demands. Proper regulation of this triad of processes during development is critical for establishing functional islets.
Collapse
|
41
|
Rodriguez-Diaz R, Abdulreda MH, Formoso AL, Gans I, Ricordi C, Berggren PO, Caicedo A. Innervation patterns of autonomic axons in the human endocrine pancreas. Cell Metab 2011; 14:45-54. [PMID: 21723503 PMCID: PMC3135265 DOI: 10.1016/j.cmet.2011.05.008] [Citation(s) in RCA: 242] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Revised: 03/30/2011] [Accepted: 05/05/2011] [Indexed: 11/18/2022]
Abstract
The autonomic nervous system regulates hormone secretion from the endocrine pancreas, the islets of Langerhans, thus impacting glucose metabolism. The parasympathetic and sympathetic nerves innervate the pancreatic islet, but the precise innervation patterns are unknown, particularly in human. Here we demonstrate that the innervation of human islets is different from that of mouse islets and does not conform to existing models of autonomic control of islet function. By visualizing axons in three dimensions and quantifying axonal densities and contacts within pancreatic islets, we found that, unlike mouse endocrine cells, human endocrine cells are sparsely contacted by autonomic axons. Few parasympathetic cholinergic axons penetrate the human islet, and the invading sympathetic fibers preferentially innervate smooth muscle cells of blood vessels located within the islet. Thus, rather than modulating endocrine cell function directly, sympathetic nerves may regulate hormone secretion in human islets by controlling local blood flow or by acting on islet regions located downstream.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Alpha cells secrete acetylcholine as a non-neuronal paracrine signal priming beta cell function in humans. Nat Med 2011; 17:888-92. [PMID: 21685896 PMCID: PMC3132226 DOI: 10.1038/nm.2371] [Citation(s) in RCA: 227] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 04/05/2011] [Indexed: 01/02/2023]
Abstract
Acetylcholine is a neurotransmitter that plays a major role in the function of the insulin secreting pancreatic beta cell1,2. Parasympathetic innervation of the endocrine pancreas, the islets of Langerhans, has been shown to provide cholinergic input to the beta cell in several species1,3,4, but the role of autonomic innervation in human beta cell function is at present unclear. Here we show that, in contrast to mouse islets, cholinergic innervation of human islets is sparse. Instead, we find that the alpha cells of the human islet provide paracrine cholinergic input to surrounding endocrine cells. Human alpha cells express the vesicular acetylcholine transporter and release acetylcholine when stimulated with kainate or a lowering in glucose concentration. Acetylcholine secretion by alpha cells in turn sensitizes the beta cell response to increases in glucose concentration. Our results demonstrate that in human islets acetylcholine is a paracrine signal that primes the beta cell to respond optimally to subsequent increases in glucose concentration. We anticipate these results to revise models about neural input and cholinergic signaling in the endocrine pancreas. Cholinergic signaling within the islet represents a potential therapeutic target in diabetes5, highlighting the relevance of this advance to future drug development.
Collapse
|
43
|
Bertram R, Sherman A, Satin LS. Electrical bursting, calcium oscillations, and synchronization of pancreatic islets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 654:261-79. [PMID: 20217502 DOI: 10.1007/978-90-481-3271-3_12] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Oscillations are an integral part of insulin secretion and are ultimately due to oscillations in the electrical activity of pancreatic beta-cells, called bursting. In this chapter we discuss islet bursting oscillations and a unified biophysical model for this multi-scale behavior. We describe how electrical bursting is related to oscillations in the intracellular Ca(2+) concentration within beta-cells and the role played by metabolic oscillations. Finally, we discuss two potential mechanisms for the synchronization of islets within the pancreas. Some degree of synchronization must occur, since distinct oscillations in insulin levels have been observed in hepatic portal blood and in peripheral blood sampling of rats, dogs, and humans. Our central hypothesis, supported by several lines of evidence, is that insulin oscillations are crucial to normal glucose homeostasis. Disturbance of oscillations, either at the level of the individual islet or at the level of islet synchronization, is detrimental and can play a major role in type 2 diabetes.
Collapse
Affiliation(s)
- Richard Bertram
- Department of Mathematics, Florida State University, Tallahassee, FL 32306, USA.
| | | | | |
Collapse
|
44
|
Olerud J, Kanaykina N, Vasylovska S, King D, Sandberg M, Jansson L, Kozlova EN. Neural crest stem cells increase beta cell proliferation and improve islet function in co-transplanted murine pancreatic islets. Diabetologia 2009; 52:2594-601. [PMID: 19823803 DOI: 10.1007/s00125-009-1544-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Accepted: 08/26/2009] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Long-term graft survival after islet transplantation to patients with type 1 diabetes is insufficient, necessitating the development of new strategies to enhance transplant viability. Here we investigated whether co-transplantation of neural crest stem cells (NCSCs) with islets improves islet survival and function in normoglycaemic and diabetic mice. METHODS Islets alone or together with NCSCs were transplanted under the kidney capsule to normoglycaemic or alloxan-induced diabetic mice. Grafts were analysed for size, proliferation, apoptosis and insulin release. In diabetic recipients blood glucose levels were examined before and after graft removal. RESULTS In mixed transplants NCSCs actively migrated and extensively associated with co-transplanted pancreatic islets. Proliferation of beta cells was markedly increased and transplants displayed improved insulin release in normoglycaemic mice compared with those receiving islet-alone transplants. Mixed grafts survived successfully and partially restored normoglycaemia in alloxan-induced diabetic mice. CONCLUSIONS/INTERPRETATION Co-grafting of NCSCs with pancreatic islets improved insulin release in mixed transplants and enhanced beta cell proliferation, resulting in increased beta cell mass. This co-transplantation model offers an opportunity to restore neural-islet interactions and improve islet functions after transplantation.
Collapse
Affiliation(s)
- J Olerud
- Department of Medical Cell Biology, Uppsala University Biomedical Center, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
45
|
Kedees MH, Grigoryan M, Guz Y, Teitelman G. Differential expression of glucagon and glucagon-like peptide 1 receptors in mouse pancreatic alpha and beta cells in two models of alpha cell hyperplasia. Mol Cell Endocrinol 2009; 311:69-76. [PMID: 19647035 PMCID: PMC2743461 DOI: 10.1016/j.mce.2009.07.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 05/26/2009] [Accepted: 07/08/2009] [Indexed: 01/06/2023]
Abstract
Glucose homeostasis is determined by a balance between insulin and glucagon, produced by beta and alpha cells of the pancreas respectively. The levels of circulating hormones is partly determined by the mass of these two endocrine cell types. However, in contrast to beta cells, the identity of the signals regulating alpha cell number is not known. Mice with a global deletion of the glucagon receptor (Gcgr-/-) and mice with ablation of prohormone convertase 2 (PC2), the enzyme involved in the conversion of proglucagon into mature glucagon, develop alpha cell hyperplasia. These observations and the fact that Gcgr-/- mice exhibit high levels of circulating glucagon-like peptide-1 (GLP-1) suggested that members of the glucagon family of peptides could be directly involved in the regulation of alpha cell number. In this study we sought to determine whether alpha cells express receptors for Glucagon (Gcgr) and/or the glucagon-like peptide-1 (GLP1r). We examined the expression of these receptors in islets of Gcgr-/-, PC2-/- mice and control littermates, in an alpha (alphaTC1/9) and in a beta (betaTC3) cell line. Gcgr was expressed exclusively by islet beta cells, but not by alpha cells, of the two lines of mice lacking glucagon signaling. Similarly, betaTC but not alphaTC cells, expressed Gcgr. The expression of GLP1r by alpha cells was determined by the genotype and age of the mice. In embryos, GLU+ cells of Gcgr+/+ mice cells express GLP1r during early development, but not in adults. In contrast, alpha cells of Gcgr-/- mice were GLP1r+ throughout life, reflecting the immature state of GLU+ cells when Gcgr is deleted. Unlike alpha cells, beta cells of all mice lines examined initiate GLP1r expression after birth. These results suggest that GLP-1 may affect the maturation of postnatal but not prenatal beta cells. In addition, they also suggest that the incretin could mediate alpha cell proliferation, inducing the development of alpha cell hyperplasia in Gcgr-/- mice.
Collapse
Affiliation(s)
- Mamdouh H Kedees
- Department of Cell Biology, State University of New York (SUNY)-Downstate Medical Center, 450 Clarkson Ave Brooklyn, NY 11203, USA
| | | | | | | |
Collapse
|
46
|
Differentiation and migration of neural crest stem cells are stimulated by pancreatic islets. Neuroreport 2009; 20:833-8. [PMID: 19421078 DOI: 10.1097/wnr.0b013e32832b8e20] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neural crest stem cells (NCSCs) migrate during embryonic development towards the endoderm-derived pancreas and the interaction between NCSCs and beta-cell progenitors is crucial for their mutual differentiation. In diabetes, loss of beta-cells or impaired beta-cell function is accompanied by nerve degeneration, which contributes to the progression of the disease. Here we show that adult pancreatic islets markedly promote differentiation of NCSCs towards neuronal phenotype in vitro and in vivo after transplantation and increase their migration towards islets. These findings indicate that pancreatic islets can be used to promote differentiation of NCSCs towards neuronal phenotype and that this in-vitro system may help elucidate interactions between NCSCs and healthy or diseased beta-cells.
Collapse
|
47
|
Fendler B, Zhang M, Satin L, Bertram R. Synchronization of pancreatic islet oscillations by intrapancreatic ganglia: a modeling study. Biophys J 2009; 97:722-9. [PMID: 19651030 PMCID: PMC2718146 DOI: 10.1016/j.bpj.2009.05.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 05/06/2009] [Accepted: 05/08/2009] [Indexed: 11/17/2022] Open
Abstract
Plasma insulin measurements from mice, rats, dogs, and humans indicate that insulin levels are oscillatory, reflecting pulsatile insulin secretion from individual islets. An unanswered question, however, is how the activity of a population of islets is coordinated to yield coherent oscillations in plasma insulin. Here, using mathematical modeling, we investigate the feasibility of a potential islet synchronization mechanism, cholinergic signaling. This hypothesis is based on well-established experimental evidence demonstrating intrapancreatic parasympathetic (cholinergic) ganglia and recent in vitro evidence that a brief application of a muscarinic agonist can transiently synchronize islets. We demonstrate using mathematical modeling that periodic pulses of acetylcholine released from cholinergic neurons is indeed able to coordinate the activity of a population of simulated islets, even if only a fraction of these are innervated. The role of islet-to-islet heterogeneity is also considered. The results suggest that the existence of cholinergic input to the pancreas may serve as a regulator of endogenous insulin pulsatility in vivo.
Collapse
Affiliation(s)
- B Fendler
- Department of Physics, Florida State University, Tallahassee, Florida, USA.
| | | | | | | |
Collapse
|
48
|
Barreto SG, Woods CM, Carati CJ, Schloithe AC, Jaya SR, Toouli J, Saccone GTP. Galanin inhibits caerulein-stimulated pancreatic amylase secretion via cholinergic nerves and insulin. Am J Physiol Gastrointest Liver Physiol 2009; 297:G333-9. [PMID: 19497960 DOI: 10.1152/ajpgi.00078.2009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pancreatic exocrine secretion is affected by galanin, but the mechanisms involved are unclear. We aimed to determine the effect and elucidate the mechanism of action of exogenous galanin on basal and stimulated pancreatic amylase secretion in vitro. The effect of galanin on basal-, carbachol-, and caerulein-stimulated amylase secretion from isolated murine pancreatic lobules was measured. Carbachol and caerulein concentration-response relationships were established. Lobules were coincubated with galanin (10(-12) M to 10(-7) M), carbachol (10(-6) M), or caerulein (10(-10) M). Lobules were preincubated with atropine (10(-5) M), tetrodotoxin (10(-5) M), hexamethonium (10(-5) M), or diazoxide (10(-7) M and 10(-4) M) for 30 min followed by incubation with caerulein (10(-10) M) alone or combined with galanin (10(-12) M). Amylase secretion was expressed as percent of total lobular amylase. Immunohistochemical studies used the antigen retrieval technique and antisera for galanin receptor (GALR) 1, 2, and 3. Carbachol and caerulein stimulated amylase secretion in a concentration-dependent manner with maximal responses of two- and 1.7-fold over control evoked at 10(-6) M and 10(-10) M, respectively. Galanin (10(-12) M) completely inhibited caerulein-stimulated amylase secretion but had no effect on carbachol-stimulated or basal secretion. Atropine and tetrodotoxin pretreatment abolished the caerulein-stimulated amylase secretion, whereas hexamethonium had no significant effect. Diazoxide significantly reduced caerulein-stimulated amylase secretion by approximately 80%. Galanin did not affect caerulein-stimulated amylase secretion in the presence of hexamethonium or diazoxide. Glucose-stimulated amylase secretion was also inhibited by galanin. Immunohistochemistry revealed islet cells labeled for GALR2. These data suggest that galanin may modulate caerulein-stimulated amylase secretion by acting on cholinergic nerves and/or islet cells possibly via GALR2 to regulate insulin release.
Collapse
Affiliation(s)
- Savio G Barreto
- Department of General and Digestive Surgery, Flinders Medical Centre and Flinders University, Adelaide, South Australia, Australia
| | | | | | | | | | | | | |
Collapse
|
49
|
Clark PB, Kavanagh K, Gage HD, Garg P, Garg S, Calles-Escandon J, Wagner JD, Morton K. Enhanced cholinergic response in pancreata of nonhuman primates with impaired glucose tolerance shown on [18F]fluorobenzyltrozamicol positron emission tomography. Diabetes Technol Ther 2009; 11:451-5. [PMID: 19580359 PMCID: PMC2902232 DOI: 10.1089/dia.2008.0113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Islet cell adaptation to insulin resistance in type 2 diabetes mellitus may be due in part to increased stimulation of beta cells by the autonomic nervous system. The parasympathetic neurotransmitter acetylcholine (ACh) mediates insulin release via M3 muscarinic receptors on islet beta cells. The vesicular ACh transporter (VAChT) receptor correlates with cholinergic activity in vivo. The positron emission tomography (PET) radiotracer (+)-4-[18F]fluorobenzyltrozamicol ([18F]FBT) binds to the VAChT receptor on presynaptic cholinergic neurons and can be quantified by PET. In this study, we utilize [18F]FBT PET to demonstrate pancreatic cholinergic activity before and after dextrose infusion in nonhuman primates with normal (NGT) and impaired (IGT) glucose tolerance. METHODS Seven adult female vervet (Chlorocebus aethiops) monkeys were maintained on an atherogenic Western diet. They were divided into two groups: four with NGT and three with IGT. Each subject underwent [18F]FBT PET twice: first, a baseline PET under fasting conditions; and second, PET under fasting conditions but after intravenous infusion of dextrose solution. Quantitative analysis of pancreatic uptake at 60 min post-injection was performed. RESULTS There was no difference in pancreatic uptake of [18F]FBT on baseline scans between the two groups. Pancreatic uptake of [18F]FBT increased in every subject after dextrose infusion (P = 0.03). On post-dextrose PET scans, pancreatic uptake of [18F]FBT was significantly higher in IGT subjects compared with NGT subjects (P = 0.03). The post-dextrose to pre-dextrose uptake ratios were higher in IGT subjects (P = 0.08). CONCLUSIONS Acute increases in pancreatic cholinergic activity in vivo were detected in the pancreata of nonhuman primates with NGT and IGT after intravenous dextrose infusion on [18F]FBT PET. In subjects with IGT, this activity was significantly higher, suggesting increased autonomic nervous system stimulation of the pancreatic islets in insulin-resistant subjects.
Collapse
Affiliation(s)
- Paige B Clark
- Department of Radiology/Nuclear Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina 27157, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Bermúdez-Silva FJ, Suárez Pérez J, Nadal A, Rodríguez de Fonseca F. The role of the pancreatic endocannabinoid system in glucose metabolism. Best Pract Res Clin Endocrinol Metab 2009; 23:87-102. [PMID: 19285263 DOI: 10.1016/j.beem.2008.10.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The endogenous cannabinoid system participates in the regulation of energy homeostasis, and this fact led to the identification of a new group of therapeutic agents for complicated obesity and diabetes. Cannabinoid receptor antagonists are now realities in clinical practice. The use of such antagonists for reducing body weight gain, lowering cholesterol and improving glucose homeostasis is based on the ability of the endocannabinoids to coordinately regulate energy homeostasis by interacting with central and peripheral targets, including adipose tissue, muscle, liver and endocrine pancreas. In this review we will analyse the presence of this system in the main cell types of the islets of Langerhans, as well as the physiological relevance of the endocannabinoids and parent acylethanolamides in hormone secretion and glucose homeostasis. We will also analyse the impact that these findings may have in clinical practice and the potential outcome of new therapeutic strategies for modulating glucose homeostasis and insulin/glucagon secretion.
Collapse
Affiliation(s)
- Francisco J Bermúdez-Silva
- Laboratorio de Medicina Regenerativa, Fundación IMABIS, Hospital Carlos Haya, Avda. Carlos Haya, Pabellón de Gobierno, sótano, 29010, Málaga, Spain.
| | | | | | | |
Collapse
|