51
|
Makhmutova M, Weitz J, Tamayo A, Pereira E, Boulina M, Almaça J, Rodriguez-Diaz R, Caicedo A. Pancreatic β-Cells Communicate With Vagal Sensory Neurons. Gastroenterology 2021; 160:875-888.e11. [PMID: 33121946 PMCID: PMC10009739 DOI: 10.1053/j.gastro.2020.10.034] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Destroying visceral sensory nerves impacts pancreatic islet function, glucose metabolism, and diabetes onset, but how islet endocrine cells interact with sensory neurons has not been studied. METHODS We characterized the anatomical pattern of pancreatic sensory innervation by combining viral tracing, immunohistochemistry, and reporter mouse models. To assess the functional interactions of β-cells with vagal sensory neurons, we recorded Ca2+ responses in individual nodose neurons in vivo while selectively stimulating β-cells with chemogenetic and pharmacologic approaches. RESULTS We found that pancreatic islets are innervated by vagal sensory axons expressing Phox2b, substance P, calcitonin-gene related peptide, and the serotonin receptor 5-HT3R. Centrally, vagal neurons projecting to the pancreas terminate in the commissural nucleus of the solitary tract. Nodose neurons responded in vivo to chemogenetic stimulation of β-cells and to pancreas infusion with serotonin, but were not sensitive to insulin. Responses to chemogenetic and pharmacologic stimulation of β-cells were blocked by a 5-HT3R antagonist and were enhanced by increasing serotonin levels in β-cells. We further confirmed directly in living pancreas slices that sensory terminals in the islet were sensitive to serotonin. CONCLUSIONS Our study establishes that pancreatic β-cells communicate with vagal sensory neurons, likely using serotonin signaling as a transduction mechanism. Serotonin is coreleased with insulin and may therefore convey information about the secretory state of β-cells via vagal afferent nerves.
Collapse
Affiliation(s)
- Madina Makhmutova
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, Florida.
| | - Jonathan Weitz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Alejandro Tamayo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Elizabeth Pereira
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida
| | - Maria Boulina
- Analytical Imaging Core Facility, Miller School of Medicine, University of Miami
| | - Joana Almaça
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Rayner Rodriguez-Diaz
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida; Program in Neuroscience, Miller School of Medicine, University of Miami, Miami, Florida; Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, Florida; Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida.
| |
Collapse
|
52
|
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
|
53
|
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: 50] [Impact Index Per Article: 10.0] [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
|
54
|
Giuliani C, Franceschi C, Luiselli D, Garagnani P, Ulijaszek S. Ecological Sensing Through Taste and Chemosensation Mediates Inflammation: A Biological Anthropological Approach. Adv Nutr 2020; 11:1671-1685. [PMID: 32647890 PMCID: PMC7666896 DOI: 10.1093/advances/nmaa078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 04/24/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022] Open
Abstract
Ecological sensing and inflammation have evolved to ensure optima between organism survival and reproductive success in different and changing environments. At the molecular level, ecological sensing consists of many types of receptors located in different tissues that orchestrate integrated responses (immune, neuroendocrine systems) to external and internal stimuli. This review describes emerging data on taste and chemosensory receptors, proposing them as broad ecological sensors and providing evidence that taste perception is shaped not only according to sense epitopes from nutrients but also in response to highly diverse external and internal stimuli. We apply a biological anthropological approach to examine how ecological sensing has been shaped by these stimuli through human evolution for complex interkingdom communication between a host and pathological and symbiotic bacteria, focusing on population-specific genetic diversity. We then focus on how these sensory receptors play a major role in inflammatory processes that form the basis of many modern common metabolic diseases such as obesity, type 2 diabetes, and aging. The impacts of human niche construction and cultural evolution in shaping environments are described with emphasis on consequent biological responsiveness.
Collapse
Affiliation(s)
- Cristina Giuliani
- Department of Biological, Geological, and Environmental Sciences (BiGeA), Laboratory of Molecular Anthropology and Centre for Genome Biology, University of Bologna, Bologna, Italy
- School of Anthropology and Museum Ethnography, University of Oxford, Oxford, United Kingdom
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Laboratory of Systems Medicine of Healthy Aging and Department of Applied Mathematics, Lobachevsky University, Nizhny Novgorod, Russia
| | - Donata Luiselli
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
- Department of Cultural Heritage (DBC), Laboratory of Ancient DNA (aDNALab), Campus of Ravenna, University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
| | - Stanley Ulijaszek
- School of Anthropology and Museum Ethnography, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
55
|
Giri TK, Roy P, Maity S. Formulation and Characterization of Solid Dispersion Containing Capsaicin for the Treatment of Diabetes. CURRENT DRUG THERAPY 2020. [DOI: 10.2174/1574885514666190916152432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Chili peppers are widely used in many cuisines as a spice, and capsaicin is
the main component. It has been reported that capsaicin acts as an antihyperglycemic agent. However,
it shows poor aqueous solubility and bioavailability.
Objective:
The is to enhance the aqueous solubility and antihyperglycemic activity of capsaicin
through solid dispersion formulation.
Methods:
Solid dispersions were prepared by the solvent evaporation method using polyethylene
glycol 6000 (PEG 6000) as a hydrophilic carrier. Polymer-drug miscibility and drug crystallinity
were characterized through the differential thermal analysis and X-ray powder patterns analysis.
Solid dispersions were evaluated for solubility, in vitro drug dissolution and in vivo animal study in
rats.
Results:
Results of x-ray powder patterns analysis showed a considerable reduction of drug crystallinity
in solid dispersion. Differential thermal analysis result revealed a complete disappearance of
capsaicin melting onset temperature in solid dispersion. From the phase solubility data, it was observed
that the aqueous solubility of capsaicin was increased with increasing concentration of PEG
6000. Solid dispersion formulation showed considerable enhancement of in vitro release of drugs in
comparison to pure capsaicin. In vivo animal study in rats shows that the solid dispersion containing
capsaicin significantly reduced the blood glucose level in comparison to the free capsaicin.
Conclusion:
Higher anti-hyperglycemic effect of capsaicin loaded solid dispersion in comparison to
the pure drug may be due to the enhancement of aqueous solubility of capsaicin. Thus, the solid
dispersion of capsaicin showed a simple approach for capsaicin delivery with improved antidiabetic
activity.
Collapse
Affiliation(s)
- Tapan Kumar Giri
- NSHM Knowledge Campus, Kolkata Group of Institutions, 124 BL Saha Road, Kolkata-700053, West Bengal, India
| | - Payel Roy
- NSHM Knowledge Campus, Kolkata Group of Institutions, 124 BL Saha Road, Kolkata-700053, West Bengal, India
| | - Subhasis Maity
- NSHM Knowledge Campus, Kolkata Group of Institutions, 124 BL Saha Road, Kolkata-700053, West Bengal, India
| |
Collapse
|
56
|
Alvarsson A, Jimenez-Gonzalez M, Li R, Rosselot C, Tzavaras N, Wu Z, Stewart AF, Garcia-Ocaña A, Stanley SA. A 3D atlas of the dynamic and regional variation of pancreatic innervation in diabetes. SCIENCE ADVANCES 2020; 6:6/41/eaaz9124. [PMID: 33036983 PMCID: PMC7557000 DOI: 10.1126/sciadv.aaz9124] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 08/27/2020] [Indexed: 05/08/2023]
Abstract
Understanding the detailed anatomy of the endocrine pancreas, its innervation, and the remodeling that occurs in diabetes can provide new insights into metabolic disease. Using tissue clearing and whole-organ imaging, we identified the 3D associations between islets and innervation. This technique provided detailed quantification of α and β cell volumes and pancreatic nerve fibers, their distribution and heterogeneity in healthy tissue, canonical mouse models of diabetes, and samples from normal and diabetic human pancreata. Innervation was highly enriched in the mouse endocrine pancreas, with regional differences. Islet nerve density was increased in nonobese diabetic mice, in mice treated with streptozotocin, and in pancreata of human donors with type 2 diabetes. Nerve contacts with β cells were preserved in diabetic mice and humans. In summary, our whole-organ assessment allows comprehensive examination of islet characteristics and their innervation and reveals dynamic regulation of islet innervation in diabetes.
Collapse
Affiliation(s)
- Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maria Jimenez-Gonzalez
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rosemary Li
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carolina Rosselot
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nikolaos Tzavaras
- The Microscopy CoRE and Advanced Bioimaging Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhuhao Wu
- Department of Cell, Developmental & Regenerative Biology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew F Stewart
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah A Stanley
- Diabetes, Obesity, and Metabolism Institute, Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
57
|
Faber CL, Deem JD, Campos CA, Taborsky GJ, Morton GJ. CNS control of the endocrine pancreas. Diabetologia 2020; 63:2086-2094. [PMID: 32894319 PMCID: PMC7983553 DOI: 10.1007/s00125-020-05204-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/07/2020] [Indexed: 12/30/2022]
Abstract
Increasing evidence suggests that, although pancreatic islets can function autonomously to detect and respond to changes in the circulating glucose level, the brain cooperates with the islet to maintain glycaemic control. Here, we review the role of the central and autonomic nervous systems in the control of the endocrine pancreas, including mechanisms whereby the brain senses circulating blood glucose levels. We also examine whether dysfunction in these systems might contribute to complications of type 1 diabetes and the pathogenesis of type 2 diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Chelsea L Faber
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer D Deem
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Carlos A Campos
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Gerald J Taborsky
- Department of Medicine, University of Washington, Seattle, WA, USA
- Veterans Affairs Puget Sound Health Care System, Department of Veterans Affairs Medical Center, Seattle, WA, USA
| | - Gregory J Morton
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, 750 Republican St, Box 358062, Seattle, WA, 98109, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
58
|
Kim M, Sisco NJ, Hilton JK, Montano CM, Castro MA, Cherry BR, Levitus M, Van Horn WD. Evidence that the TRPV1 S1-S4 membrane domain contributes to thermosensing. Nat Commun 2020; 11:4169. [PMID: 32820172 PMCID: PMC7441067 DOI: 10.1038/s41467-020-18026-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/30/2020] [Indexed: 01/14/2023] Open
Abstract
Sensing and responding to temperature is crucial in biology. The TRPV1 ion channel is a well-studied heat-sensing receptor that is also activated by vanilloid compounds, including capsaicin. Despite significant interest, the molecular underpinnings of thermosensing have remained elusive. The TRPV1 S1-S4 membrane domain couples chemical ligand binding to the pore domain during channel gating. Here we show that the S1-S4 domain also significantly contributes to thermosensing and couples to heat-activated gating. Evaluation of the isolated human TRPV1 S1-S4 domain by solution NMR, far-UV CD, and intrinsic fluorescence shows that this domain undergoes a non-denaturing temperature-dependent transition with a high thermosensitivity. Further NMR characterization of the temperature-dependent conformational changes suggests the contribution of the S1-S4 domain to thermosensing shares features with known coupling mechanisms between this domain with ligand and pH activation. Taken together, this study shows that the TRPV1 S1-S4 domain contributes to TRPV1 temperature-dependent activation.
Collapse
Affiliation(s)
- Minjoo Kim
- School of Molecular Sciences, Arizona State University, 551 E. University Drive, Tempe, AZ, 85287, USA
- The Biodesign Institute Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA
| | - Nicholas J Sisco
- School of Molecular Sciences, Arizona State University, 551 E. University Drive, Tempe, AZ, 85287, USA
- The Biodesign Institute Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA
| | - Jacob K Hilton
- School of Molecular Sciences, Arizona State University, 551 E. University Drive, Tempe, AZ, 85287, USA
- The Biodesign Institute Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA
| | - Camila M Montano
- The Biodesign Institute Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA
| | - Manuel A Castro
- School of Molecular Sciences, Arizona State University, 551 E. University Drive, Tempe, AZ, 85287, USA
| | - Brian R Cherry
- The Magnetic Resonance Research Center, Arizona State University, Tempe, AZ, 85287, USA
| | - Marcia Levitus
- School of Molecular Sciences, Arizona State University, 551 E. University Drive, Tempe, AZ, 85287, USA
- The Biodesign Institute Center for Single Molecule Biophysics, Arizona State University, Tempe, AZ, 85287, USA
| | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University, 551 E. University Drive, Tempe, AZ, 85287, USA.
- The Biodesign Institute Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
59
|
Christoffersson G, Ratliff SS, von Herrath MG. Interference with pancreatic sympathetic signaling halts the onset of diabetes in mice. SCIENCE ADVANCES 2020; 6:6/35/eabb2878. [PMID: 33052874 PMCID: PMC7531904 DOI: 10.1126/sciadv.abb2878] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/13/2020] [Indexed: 05/04/2023]
Abstract
The notably lobular distribution of immune lesions in type 1 diabetes (T1D) has been hypothesized to be the result of innervation within the pancreas. To investigate whether neuroimmune interactions could explain this phenomenon, we explored the impact of sympathetic signaling in the RIP-LCMV-GP mouse model of autoimmune diabetes. In this model, the CD8+ T cell attack on β cells replicates a key pathogenic feature of human T1D. We found that inhibition of α1 adrenoceptors, ablation of sympathetic nerves, and surgical denervation all had a protective effect in this model, without affecting the systemic presence of β cell-reactive CD8+ T cells. In vivo multiphoton imaging revealed a local effect within pancreatic islets including limited infiltration of both macrophages and β cell-specific CD8+ T cells. Islet-resident macrophages expressed adrenoceptors and were responsive to catecholamines. Islet macrophages may therefore constitute a pivotal neuroimmune signaling relay and could be a target for future interventions in T1D.
Collapse
Affiliation(s)
- Gustaf Christoffersson
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA.
- Department of Medical Cell Biology, Uppsala University, Uppsala 75237, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala 75237, Sweden
| | | | - Matthias G von Herrath
- La Jolla Institute for Immunology, La Jolla, CA 92037, USA.
- Novo Nordisk Research Center, Seattle, WA 98109, USA
| |
Collapse
|
60
|
Distribution of nerve fibers and nerve-immune cell association in mouse spleen revealed by immunofluorescent staining. Sci Rep 2020; 10:9850. [PMID: 32555231 PMCID: PMC7300136 DOI: 10.1038/s41598-020-66619-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/21/2020] [Indexed: 02/05/2023] Open
Abstract
The central nervous system regulates the immune system through the secretion of hormones from the pituitary gland and other endocrine organs, while the peripheral nervous system (PNS) communicates with the immune system through local nerve-immune cell interactions, including sympathetic/parasympathetic (efferent) and sensory (afferent) innervation to lymphoid tissue/organs. However, the precise mechanisms of this bi-directional crosstalk of the PNS and immune system remain mysterious. To study this kind of bi-directional crosstalk, we performed immunofluorescent staining of neurofilament and confocal microscopy to reveal the distribution of nerve fibers and nerve-immune cell associations inside mouse spleen. Our study demonstrates (i) extensive nerve fibers in all splenic compartments including the splenic nodules, periarteriolar lymphoid sheath, marginal zones, trabeculae, and red pulp; (ii) close associations of nerve fibers with blood vessels (including central arteries, marginal sinuses, penicillar arterioles, and splenic sinuses); (iii) close associations of nerve fibers with various subsets of dendritic cells, macrophages (Mac1+ and F4/80+), and lymphocytes (B cells, T helper cells, and cytotoxic T cells). Our data concerning the extensive splenic innervation and nerve-immune cell communication will enrich our knowledge of the mechanisms through which the PNS affects the cellular- and humoral-mediated immune responses in healthy and infectious/non-infectious states.
Collapse
|
61
|
Lázár BA, Jancsó G, Sántha P. Modulation of Sensory Nerve Function by Insulin: Possible Relevance to Pain, Inflammation and Axon Growth. Int J Mol Sci 2020; 21:E2507. [PMID: 32260335 PMCID: PMC7177741 DOI: 10.3390/ijms21072507] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/16/2022] Open
Abstract
Insulin, besides its pivotal role in energy metabolism, may also modulate neuronal processes through acting on insulin receptors (InsRs) expressed by neurons of both the central and the peripheral nervous system. Recently, the distribution and functional significance of InsRs localized on a subset of multifunctional primary sensory neurons (PSNs) have been revealed. Systematic investigations into the cellular electrophysiology, neurochemistry and morphological traits of InsR-expressing PSNs indicated complex functional interactions among specific ion channels, proteins and neuropeptides localized in these neurons. Quantitative immunohistochemical studies have revealed disparate localization of the InsRs in somatic and visceral PSNs with a dominance of InsR-positive neurons innervating visceral organs. These findings suggested that visceral spinal PSNs involved in nociceptive and inflammatory processes are more prone to the modulatory effects of insulin than somatic PSNs. Co-localization of the InsR and transient receptor potential vanilloid 1 (TRPV1) receptor with vasoactive neuropeptides calcitonin gene-related peptide and substance P bears of crucial importance in the pathogenesis of inflammatory pathologies affecting visceral organs, such as the pancreas and the urinary bladder. Recent studies have also revealed significant novel aspects of the neurotrophic propensities of insulin with respect to axonal growth, development and regeneration.
Collapse
Affiliation(s)
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, H-6720 Szeged, Hungary; (G.J.); (P.S.)
| | - Péter Sántha
- Department of Physiology, University of Szeged, H-6720 Szeged, Hungary; (G.J.); (P.S.)
| |
Collapse
|
62
|
Islam MS. Molecular Regulations and Functions of the Transient Receptor Potential Channels of the Islets of Langerhans and Insulinoma Cells. Cells 2020; 9:cells9030685. [PMID: 32168890 PMCID: PMC7140661 DOI: 10.3390/cells9030685] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 12/17/2022] Open
Abstract
Insulin secretion from the β-cells of the islets of Langerhans is triggered mainly by nutrients such as glucose, and incretin hormones such as glucagon-like peptide-1 (GLP-1). The mechanisms of the stimulus-secretion coupling involve the participation of the key enzymes that metabolize the nutrients, and numerous ion channels that mediate the electrical activity. Several members of the transient receptor potential (TRP) channels participate in the processes that mediate the electrical activities and Ca2+ oscillations in these cells. Human β-cells express TRPC1, TRPM2, TRPM3, TRPM4, TRPM7, TRPP1, TRPML1, and TRPML3 channels. Some of these channels have been reported to mediate background depolarizing currents, store-operated Ca2+ entry (SOCE), electrical activity, Ca2+ oscillations, gene transcription, cell-death, and insulin secretion in response to stimulation by glucose and GLP1. Different channels of the TRP family are regulated by one or more of the following mechanisms: activation of G protein-coupled receptors, the filling state of the endoplasmic reticulum Ca2+ store, heat, oxidative stress, or some second messengers. This review briefly compiles our current knowledge about the molecular mechanisms of regulations, and functions of the TRP channels in the β-cells, the α-cells, and some insulinoma cell lines.
Collapse
Affiliation(s)
- Md. Shahidul Islam
- Karolinska Institutet, Department of Clinical Science and Education, Södersjukhuset, Research Center, 5th floor, SE-118 83 Stockholm, Sweden;
- Department of Emergency Care and Internal Medicine, Uppsala University Hospital, Uppsala University, SE-751 85 Uppsala, Sweden
| |
Collapse
|
63
|
Zhong B, Ma S, Wang DH. TRPV1 Mediates Glucose-induced Insulin Secretion Through Releasing Neuropeptides. In Vivo 2020; 33:1431-1437. [PMID: 31471389 DOI: 10.21873/invivo.11621] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 06/28/2019] [Accepted: 07/03/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND/AIM Transient receptor potential vanilloid 1 (TRPV1)-expressing sensory nerves innervate the pancreatic islets. Sensory neuropeptides, including calcitonin gene-related peptide (CGRP) and substance P (SP), participate in insulin secretion. This study aimed to investigate the role of TRPV1 in glucose-induced insulin secretion. MATERIALS AND METHODS TRPV1-/- and wild-type (WT) mice were fed a normal diet for 24 weeks. Glucose tolerance and insulin secretion were measured at the end of the experiments. RESULTS TRPV1-/- mice had greater impairments in glucose tolerance and higher decrease in glucose-induced insulin secretion than WT mice. Capsaicin (a TRPV1 agonist) increased insulin secretion in WT, but not in TRPV1-/- mice. Glucose-induced insulin secretion was blunted in TRPV1-/- mice, and was attenuated by AMG9810 (a TRPV1 inhibitor), CGRP8-37 (a CGRP receptor antagonist), or RP67580 (a NK-1 receptor antagonist) in WT mice. Glucose-induced SP and CGRP release from WT pancreas was higher than that from TRPV1-/- pancreas. CONCLUSION TRPV1 mediates glucose-induced insulin secretion likely through CGRP and SP release.
Collapse
Affiliation(s)
- Beihua Zhong
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, U.S.A
| | - Shuangtao Ma
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, U.S.A
| | - Donna H Wang
- Division of Nanomedicine and Molecular Intervention, Department of Medicine, Michigan State University, East Lansing, MI, U.S.A. .,Neuroscience Program, Michigan State University, East Lansing, MI, U.S.A.,Cell & Molecular Biology Program, Michigan State University, East Lansing, MI, U.S.A
| |
Collapse
|
64
|
Jayasimhan A, Mariño E. Dietary SCFAs, IL-22, and GFAP: The Three Musketeers in the Gut-Neuro-Immune Network in Type 1 Diabetes. Front Immunol 2019; 10:2429. [PMID: 31736937 PMCID: PMC6828936 DOI: 10.3389/fimmu.2019.02429] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/30/2019] [Indexed: 01/13/2023] Open
Abstract
Microbial metabolites have a profound effect on the development of type 1 diabetes (T1D). The cross-talk between the gut microbiota, the nervous system, and immune system is necessary to establish and maintain immune and gut tolerance. As quoted by Hippocrates, "All disease begins in the gut." Although this has been recognized for 2,000 years, the connection between the gut and autoimmune T1D is not yet well-understood. Here, we outline new advances supported by our research and others that have contributed to elucidate the impact of microbial metabolites on the physiology of the pancreas and the gut through their remarkable effect on the immune and nervous system. Among many of the mechanisms involved in the gut-beta-cell-immune cross-talk, glial fibrillary acidic protein (GFAP)-expressing cells are critical players in the development of invasive insulitis. Besides, this review reveals a novel mechanism for microbial metabolites by stimulating IL-22, an essential cytokine for gut homeostasis and beta-cell survival. The close connections between the gut and the pancreas are highlighted through our review as microbial metabolites recirculate through the whole body and intimately react with the nervous system, which controls essential disorders associated with diabetes. As such, we discuss the mechanisms of action of microbial metabolites or short-chain fatty acids (SCFAs), IL-22, and GFAP on beta-cells, gut epithelial cells, neurons, and glial cells via metabolite sensing receptors or through epigenetic effects. The fine-tuned gut-neuro-immune network may be profoundly affected by SCFA deficiency related to dysbiosis and diet alterations at very early stages of the initiation of the disease. Thus, dampening the initial immune response or preventing the perpetuation of the immune response by maintaining the integrity of the gut is among the alternative approaches to prevent T1D.
Collapse
Affiliation(s)
- Abhirup Jayasimhan
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Eliana Mariño
- Infection and Immunity Program, Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
65
|
Hu D, Nicholls PK, Claus M, Wu Y, Shi Z, Greene WK, Ma B. Immunofluorescence characterization of innervation and nerve-immune cell interactions in mouse lymph nodes. Eur J Histochem 2019; 63. [PMID: 31631646 PMCID: PMC6802453 DOI: 10.4081/ejh.2019.3059] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/30/2019] [Indexed: 12/22/2022] Open
Abstract
The peripheral nervous system communicates specifically with the immune system via local interactions. These interactions include the “hardwiring” of sympathetic/ parasympathetic (efferent) and sensory nerves (afferent) to primary (e.g., thymus and bone marrow) and secondary (e.g., lymph node, spleen, and gut-associated lymphoid tissue) lymphoid tissue/organs. To gain a better understanding of this bidirectional interaction/crosstalk between the two systems, we have investigated the distribution of nerve fibres and PNS-immune cell associations in situ in the mouse lymph node by using immunofluorescent staining and confocal microscopy/ three-dimensional reconstruction. Our results demonstrate: i) the presence of extensive nerve fibres in all compartments (including B cell follicles) in the mouse lymph node; ii) close contacts/ associations of nerve fibres with blood vessels (including high endothelial venules) and lymphatic vessels/sinuses; iii) close contacts/associations of nerve fibres with various subsets of dendritic cells (e.g., B220+CD11c+, CD4+CD11c+, CD8a+ CD11c+, and Mac1+CD11c+), Mac1+ macrophages, and B/T lymphocytes. Our novel findings concerning the innervation and nerve-immune cell interactions inside the mouse lymph node should greatly facilitate our understanding of the effects that the peripheral nervous system has on cellular- and humoral-mediated immune responses or vice versa in health and disease.
Collapse
Affiliation(s)
- Dailun Hu
- Clinical College, Hebei Medical University, Shijiazhuang.
| | | | | | | | | | | | | |
Collapse
|
66
|
Dhukhwa A, Bhatta P, Sheth S, Korrapati K, Tieu C, Mamillapalli C, Ramkumar V, Mukherjea D. Targeting Inflammatory Processes Mediated by TRPVI and TNF-α for Treating Noise-Induced Hearing Loss. Front Cell Neurosci 2019; 13:444. [PMID: 31632242 PMCID: PMC6786284 DOI: 10.3389/fncel.2019.00444] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 09/18/2019] [Indexed: 12/20/2022] Open
Abstract
Noise trauma is the most common cause of hearing loss in adults. There are no known FDA approved drugs for prevention or rescue of noise-induced hearing loss (NIHL). In this study, we provide evidence that implicates stress signaling molecules (TRPV1, NOX3, and TNF-α) in NIHL. Furthermore, we provide evidence that inhibiting any one of these moieties can prevent and treat NIHL when administered within a window period. Hearing loss induced by loud noise is associated with the generation of reactive oxygen species (ROS), increased calcium (Ca2+) in the endolymph and hair cells, and increased inflammation in the cochlea. Increased (Ca2+) and ROS activity persists for several days after traumatic noise exposure (NE). Chronic increases in (Ca2+) and ROS have been shown to increase inflammation and apoptosis in various tissue. However, the precise role of Ca2+ up-regulation and the resulting inflammation causing a positive feedback loop in the noise-exposed cochlea to generate sustained toxic amounts of Ca2+ are unknown. Here we show cochlear TRPV1 dysregulation is a key step in NIHL, and that inflammatory TNF-α cytokine-mediated potentiation of TRPV1 induced Ca2+ entry is an essential mechanism of NIHL. In the Wistar rat model, noise produces an acute (within 48 h) and a chronic (within 21 days) increase in cochlear gene expression of TRPV1, NADPH oxidase 3 (NOX3) and pro-inflammatory mediators such as tumor necrosis factor-α (TNF-α) and cyclooxygenase-2 (COX2). Additionally, we also show that H2O2 (100 μM) produces a robust increase in Ca2+ entry in cell cultures which is enhanced by TNF-α via the TRPV1 channel and which involves ERK1/2 phosphorylation. Mitigation of NIHL could be achieved by using capsaicin (TRPV1 agonist that rapidly desensitizes TRPV1. This mechanism is used in the treatment of pain in diabetic peripheral neuropathy) pretreatment or by inhibition of TNF-α with Etanercept (ETA), administered up to 7 days prior to NE or within 24 h of noise. Our results demonstrate the importance of the synergistic interaction between TNF-α and TRPV1 in the cochlea and suggest that these are important therapeutic targets for treating NIHL.
Collapse
Affiliation(s)
- Asmita Dhukhwa
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Puspanjali Bhatta
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Sandeep Sheth
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, United States
| | - Krishi Korrapati
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Coral Tieu
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Chaitanya Mamillapalli
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Vickram Ramkumar
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, United States
| | - Debashree Mukherjea
- Department of Otolaryngology, Southern Illinois University School of Medicine, Springfield, IL, United States
| |
Collapse
|
67
|
Laumet G, Ma J, Robison AJ, Kumari S, Heijnen CJ, Kavelaars A. T Cells as an Emerging Target for Chronic Pain Therapy. Front Mol Neurosci 2019; 12:216. [PMID: 31572125 PMCID: PMC6749081 DOI: 10.3389/fnmol.2019.00216] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/26/2019] [Indexed: 11/13/2022] Open
Abstract
The immune system is critically involved in the development and maintenance of chronic pain. However, T cells, one of the main regulators of the immune response, have only recently become a focus of investigations on chronic pain pathophysiology. Emerging clinical data suggest that patients with chronic pain have a different phenotypic profile of circulating T cells compared to controls. At the preclinical level, findings on the function of T cells are mixed and differ between nerve injury, chemotherapy, and inflammatory models of persistent pain. Depending on the type of injury, the subset of T cells and the sex of the animal, T cells may contribute to the onset and/or the resolution of pain, underlining T cells as a major player in the transition from acute to chronic pain. Specific T cell subsets release mediators such as cytokines and endogenous opioid peptides that can promote, suppress, or even resolve pain. Inhibiting the pain-promoting functions of T cells and/or enhancing the beneficial effects of pro-resolution T cells may offer new disease-modifying strategies for the treatment of chronic pain, a critical need in view of the current opioid crisis.
Collapse
Affiliation(s)
- Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, United States.,Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jiacheng Ma
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Susmita Kumari
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
68
|
Crosson T, Roversi K, Balood M, Othman R, Ahmadi M, Wang JC, Seadi Pereira PJ, Tabatabaei M, Couture R, Eichwald T, Latini A, Prediger RD, Rangachari M, Seehus CR, Foster SL, Talbot S. Profiling of how nociceptor neurons detect danger - new and old foes. J Intern Med 2019; 286:268-289. [PMID: 31282104 DOI: 10.1111/joim.12957] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The host evolves redundant mechanisms to preserve physiological processing and homeostasis. These functions range from sensing internal and external threats, creating a memory of the insult and generating reflexes, which aim to resolve inflammation. Impairment in such functioning leads to chronic inflammatory diseases. By interacting through a common language of ligands and receptors, the immune and sensory nervous systems work in concert to accomplish such protective functions. Whilst this bidirectional communication helps to protect from danger, it can contribute to disease pathophysiology. Thus, the somatosensory nervous system is anatomically positioned within primary and secondary lymphoid tissues and mucosa to modulate immunity directly. Upstream of this interplay, neurons detect danger, which prompts the release of neuropeptides initiating (i) defensive reflexes (ranging from withdrawal response to coughing) and (ii) chemotaxis, adhesion and local infiltration of immune cells. The resulting outcome of such neuro-immune interplay is still ill-defined, but consensual findings start to emerge and support neuropeptides not only as blockers of TH 1-mediated immunity but also as drivers of TH 2 immune responses. However, the modalities detected by nociceptors revealed broader than mechanical pressure and temperature sensing and include signals as various as cytokines and pathogens to immunoglobulins and even microRNAs. Along these lines, we aggregated various dorsal root ganglion sensory neuron expression profiling datasets supporting such wide-ranging sensing capabilities to help identifying new danger detection modalities of these cells. Thus, revealing unexpected aspects of nociceptor neuron biology might prompt the identification of novel drivers of immunity, means to resolve inflammation and strategies to safeguard homeostasis.
Collapse
Affiliation(s)
- T Crosson
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - K Roversi
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada.,Departamento de Farmacologia Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - M Balood
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada.,Axe Neurosciences, Centre de recherche du CHU, Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - R Othman
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - M Ahmadi
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - J-C Wang
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada.,Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | - M Tabatabaei
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - R Couture
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| | - T Eichwald
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - A Latini
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - R D Prediger
- Departamento de Farmacologia Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - M Rangachari
- Axe Neurosciences, Centre de recherche du CHU, Université Laval, Québec, QC, Canada.,Département de Médecine Moléculaire, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - C R Seehus
- FM Kirby Neurobiology Center, Children's Hospital, Boston, MA, USA
| | - S L Foster
- Depression Clinical Research Program, Massachusetts General Hospital, Boston, MA, USA
| | - S Talbot
- From the, Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
69
|
Dhakal S, Lee Y. Transient Receptor Potential Channels and Metabolism. Mol Cells 2019; 42:569-578. [PMID: 31446746 PMCID: PMC6715338 DOI: 10.14348/molcells.2019.0007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 07/27/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Transient receptor potential (TRP) channels are nonselective cationic channels, conserved among flies to humans. Most TRP channels have well known functions in chemosensation, thermosensation, and mechanosensation. In addition to being sensing environmental changes, many TRP channels are also internal sensors that help maintain homeostasis. Recent improvements to analytical methods for genomics and metabolomics allow us to investigate these channels in both mutant animals and humans. In this review, we discuss three aspects of TRP channels, which are their role in metabolism, their functional characteristics, and their role in metabolic syndrome. First, we introduce each TRP channel superfamily and their particular roles in metabolism. Second, we provide evidence for which metabolites TRP channels affect, such as lipids or glucose. Third, we discuss correlations between TRP channels and obesity, diabetes, and mucolipidosis. The cellular metabolism of TRP channels gives us possible therapeutic approaches for an effective prophylaxis of metabolic syndromes.
Collapse
Affiliation(s)
- Subash Dhakal
- Department of Bio and Fermentation Convergence Technology, Kookmin University, BK21 PLUS Project, Seoul 02707,
Korea
| | - Youngseok Lee
- Department of Bio and Fermentation Convergence Technology, Kookmin University, BK21 PLUS Project, Seoul 02707,
Korea
| |
Collapse
|
70
|
Liu J, Du J, Wang Y. CDK5 inhibits the clathrin-dependent internalization of TRPV1 by phosphorylating the clathrin adaptor protein AP2μ2. Sci Signal 2019; 12:12/585/eaaw2040. [PMID: 31186372 DOI: 10.1126/scisignal.aaw2040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Transient receptor potential vanilloid 1 (TRPV1), a nonselective, ligand-gated cation channel, responds to multiple noxious stimuli and is targeted by many kinases that influence its trafficking and activity. Studies on the internalization of TRPV1 have mainly focused on that induced by capsaicin or other agonists. Here, we report that constitutive internalization of TRPV1 occurred in a manner dependent on clathrin, dynamin, and adaptor protein complex 2 (AP2). The μ2 subunit of AP2 (AP2μ2) interacted directly with TRPV1 and was required for its constitutive internalization. Cyclin-dependent kinase 5 (CDK5) phosphorylated AP2μ2 at Ser45, which reduced the interaction between TRPV1 and AP2μ2, leading to decreased TRPV1 internalization. Intrathecal delivery of a cell-penetrating fusion peptide corresponding to the Cdk5 phosphorylation site in AP2μ2, which competed with AP2μ2 for phosphorylation by Cdk5, increased the abundance of TRPV1 on the surface of dorsal root ganglion neurons and reduced complete Freund's adjuvant (CFA)-induced inflammatory thermal hyperalgesia in rats. In addition to describing a mechanism of TRPV1 constitutive internalization and its inhibition by CDK5, these findings demonstrate that CDK5 promotes inflammatory thermal hyperalgesia by reducing TRPV1 internalization, providing previously unidentified insights into the search for drug targets to treat pain.
Collapse
Affiliation(s)
- Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, China.,Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, the Key Laboratory for Neuroscience of the Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| | - Junxia Du
- College of Biological Science and Engineering, Xingtai University, Xingtai 054001, Hebei Province, China
| | - Yun Wang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, the Key Laboratory for Neuroscience of the Ministry of Education/National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China. .,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| |
Collapse
|
71
|
Alexandre-Heymann L, Mallone R, Boitard C, Scharfmann R, Larger E. Structure and function of the exocrine pancreas in patients with type 1 diabetes. Rev Endocr Metab Disord 2019; 20:129-149. [PMID: 31077020 DOI: 10.1007/s11154-019-09501-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the last 10 years, several studies have shown that the pancreas of patients with type 1 diabetes (T1D), and even of subjects at risk for T1D, was smaller than the pancreas from healthy subjects. This arose the question of the relationships between the endocrine and exocrine parts of the pancreas in T1D pathogenesis. Our review underlines that histological anomalies of the exocrine pancreas are common in patients with T1D: intralobular and interacinar fibrosis, acinar atrophy, fatty infiltration, leucocytic infiltration, and pancreatic arteriosclerosis are all frequent observations. Moreover, 25% to 75% of adult patients with T1D present with pancreatic exocrine dysfunction. Our review summarizes the putative causal factors for these structural and functional anomalies, including: 1/ alterations of insulin, glucagon, somatostatin and pancreatic polypeptide secretion, 2/ global pancreatic inflammation 3/ autoimmunity targeting the exocrine pancreas, 4/ vascular and neural abnormalities, and 5/ the putative involvement of pancreatic stellate cells. These observations have also given rise to new theories on T1D: the primary event of T1D pathogenesis could be non-specific, e.g bacterial or viral or chemical, resulting in global pancreatic inflammation, which in turn could cause beta-cell predominant destruction by the immune system. Finally, this review emphasizes that it is advisable to evaluate pancreatic exocrine function in patients with T1D presenting with gastro-intestinal complaints, as a clinical trial has shown that pancreatic enzymes replacement therapy can reduce the frequency of hypoglycemia and thus might improve quality of life in subjects with T1D and exocrine failure.
Collapse
Affiliation(s)
- Laure Alexandre-Heymann
- Service de Diabétologie, Hôpital Cochin, 123 boulevard de Port-Royal, 75014, Paris, France
- Département Hospitalo Universitaire, INSERM U 1016, Université Paris Descartes, Paris, France
| | - Roberto Mallone
- Service de Diabétologie, Hôpital Cochin, 123 boulevard de Port-Royal, 75014, Paris, France
- Département Hospitalo Universitaire, INSERM U 1016, Université Paris Descartes, Paris, France
| | - Christian Boitard
- Service de Diabétologie, Hôpital Cochin, 123 boulevard de Port-Royal, 75014, Paris, France
- Département Hospitalo Universitaire, INSERM U 1016, Université Paris Descartes, Paris, France
| | - Raphaël Scharfmann
- Service de Diabétologie, Hôpital Cochin, 123 boulevard de Port-Royal, 75014, Paris, France
- Département Hospitalo Universitaire, INSERM U 1016, Université Paris Descartes, Paris, France
| | - Etienne Larger
- Service de Diabétologie, Hôpital Cochin, 123 boulevard de Port-Royal, 75014, Paris, France.
- Département Hospitalo Universitaire, INSERM U 1016, Université Paris Descartes, Paris, France.
| |
Collapse
|
72
|
Jayasimhan A, Ellis DP, Ziegler AI, Slattery RM. Pancreatic ductal cell antigens are important in the development of invasive insulitis in Non-Obese Diabetic mice. J Neuroimmunol 2019; 327:1-9. [PMID: 30685070 DOI: 10.1016/j.jneuroim.2019.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 01/12/2023]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease in which insulin producing beta cells of the pancreas are selectively destroyed. Glial Fibrillary Acidic Protein (GFAP) expressed in peri-islet Schwann cells (pSCs) and in the ductal cells of the pancreas is one of the candidate autoantigens for T1D. Immune responses to GFAP expressing cell types precede the islet autoimmunity in Non-Obese Diabetic (NOD) mice. By removing MHC class I from GFAP expressing cell types, we tested the role of autoantigens presented by these cell types in the development of invasive insulitis. Our findings indicate that antigens expressed by pancreatic ductal cells are important in the development of invasive insulitis in NOD mice.
Collapse
Affiliation(s)
- Abhirup Jayasimhan
- Department of Immunology and Pathology, Monash University, Melbourne, Australia.
| | - Darcy P Ellis
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Alexandra I Ziegler
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Robyn M Slattery
- Department of Immunology and Pathology, Monash University, Melbourne, Australia
| |
Collapse
|
73
|
Molinas AJR, Desmoulins LD, Hamling BV, Butcher SM, Anwar IJ, Miyata K, Enix CL, Dugas CM, Satou R, Derbenev AV, Zsombok A. Interaction between TRPV1-expressing neurons in the hypothalamus. J Neurophysiol 2019; 121:140-151. [PMID: 30461371 PMCID: PMC6383661 DOI: 10.1152/jn.00004.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 10/29/2018] [Accepted: 11/14/2018] [Indexed: 02/08/2023] Open
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel expressed in the peripheral and central nervous systems. TRPV1-dependent mechanisms take part in a wide range of physiological and pathophysiological pathways including the regulation of homeostatic functions. TRPV1 expression in the hypothalamus has been described as well as evidence that TRPV1-dependent excitatory inputs to hypothalamic preautonomic neurons are diminished in diabetic conditions. Here we aimed to determine the functional expression of TRPV1 in two hypothalamic nuclei known to be involved in the central control of metabolism and to test the hypothesis that TRPV1-expressing neurons receive TRPV1-expressing inputs. A mouse model (TRPV1Cre/tdTom) was generated to identify TRPV1-expressing cells and determine the cellular properties of TRPV1-expressing neurons in adult mice. Our study demonstrated the functional expression of TRPV1 in the dorsomedial hypothalamic nucleus and paraventricular nucleus in adult mice. Our findings revealed that a subset of TRPV1Cre/tdTom neurons receive TRPV1-expressing excitatory inputs, indicating direct interaction between TRPV1-expressing neurons. In addition, astrocytes likely play a role in the modulation of TRPV1-expressing neurons. In summary, this study identified specific hypothalamic regions where TRPV1 is expressed and functional in adult mice and the existence of direct connections between TRPV1Cre/tdTom neurons. NEW & NOTEWORTHY Transient receptor potential vanilloid type 1 (TRPV1) is expressed in the hypothalamus, and TRPV1-dependent regulation of preautonomic neurons is decreased in hyperglycemic conditions. Our study demonstrated functional expression of TRPV1 in two hypothalamic nuclei involved in the control of energy homeostasis. Our results also revealed that a subset of TRPV1-expressing neurons receive TRPV1-expressing excitatory inputs. These findings suggest direct interaction between TRPV1-expressing neurons.
Collapse
Affiliation(s)
- Adrien J R Molinas
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Lucie D Desmoulins
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Brooke V Hamling
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
- Neuroscience Program, Brain Institute, Tulane University , New Orleans, Louisiana
| | - Sierra M Butcher
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
- Neuroscience Program, Brain Institute, Tulane University , New Orleans, Louisiana
| | - Imran J Anwar
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Kayoko Miyata
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Courtney L Enix
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Courtney M Dugas
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Ryousuke Satou
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
| | - Andrei V Derbenev
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
- Neuroscience Program, Brain Institute, Tulane University , New Orleans, Louisiana
| | - Andrea Zsombok
- Department of Physiology, School of Medicine, Tulane University , New Orleans, Louisiana
- Neuroscience Program, Brain Institute, Tulane University , New Orleans, Louisiana
| |
Collapse
|
74
|
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: 1.9] [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
|
75
|
Zhang N, Gao D, Liu Y, Ji S, Sha L. Effects of Neuropeptide Substance P on Proliferation and β-Cell Differentiation of Adult Pancreatic Ductal Cells. Front Neurosci 2018; 12:806. [PMID: 30455626 PMCID: PMC6230717 DOI: 10.3389/fnins.2018.00806] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/16/2018] [Indexed: 01/23/2023] Open
Abstract
Purpose: The pancreas is innervated by sensory nerves, parasympathetic and sympathetic nerves. The classical neurotransmitters, acetylcholine and noradrenaline, and some kind of neuropeptides are contained in the terminals of these nerves. Neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) co-released from the primary sensory fibers have been identified as the key neurotransmitters in pancreas. Pancreatic ductal epithelium cells are one of the important sources of the pancreatic islet β-cell neogenesis. We hypothesized that SP and CGRP might play a role on proliferation of ductal cells and differentiation of ductal cells toward the β-cell neogenesis. Methods: Primary ductal cells of rat pancreas at the third passage (P3) were used. The identification of P3 cells were confirmed with flow cytometry analysis and immunostaining by CK19 (the ductal cell marker). Proliferation of ductal cells was verified by CCK-8 assay and Ki67 immunostaining. Differentiation of ductal cells was determined with immunostaining and flow cytometry. Possible mechanism was explored by testing the key proteins of Wnt signaling using Western blot analysis. Results: Our data showed that SP but not CGRP promoted proliferation of ductal cells. Moreover, NK-1 receptor antagonist L-703,606 blocked the SP-induced stimulation of proliferation. The results of Western blot analysis showed that L-703,606 attenuated the effects of substance P on NK1R, GSK-3β, and β-catenin expression. However, SP did not directly induce the differentiation of ductal cells into β-cells, and did not promote the progression of ductal cells to differentiate into more insulin-produced cells in induction medium. Conclusions: These findings suggested that SP but not CGRP promoted proliferation of adult pancreatic ductal cells. SP promoted proliferation of ductal cells but not differentiation into β-cells. NK1R and Wnt signaling pathway might be involved in the mechanism of promoting the proliferation of ductal cells by SP. Findings in this study indicated the lack of SP might be a possible indicator for the initial of diabetes. SP could also be used as a drug candidate for the treatment of diabetes.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Di Gao
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Yudan Liu
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Sihan Ji
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Lei Sha
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
76
|
Lázár BA, Jancsó G, Pálvölgyi L, Dobos I, Nagy I, Sántha P. Insulin Confers Differing Effects on Neurite Outgrowth in Separate Populations of Cultured Dorsal Root Ganglion Neurons: The Role of the Insulin Receptor. Front Neurosci 2018; 12:732. [PMID: 30364236 PMCID: PMC6191510 DOI: 10.3389/fnins.2018.00732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023] Open
Abstract
Apart from its pivotal role in the regulation of carbohydrate metabolism, insulin exerts important neurotrophic and neuromodulator effects on dorsal root ganglion (DRG) neurons. The neurite outgrowth-promoting effect is one of the salient features of insulin's action on cultured DRG neurons. Although it has been established that a significant population of DRG neurons express the insulin receptor (InsR), the significance of InsR expression and the chemical phenotype of DRG neurons in relation to the neurite outgrowth-promoting effect of insulin has not been studied. Therefore, in this study by using immunohistochemical and quantitative stereological methods we evaluated the effect of insulin on neurite outgrowth of DRG neurons of different chemical phenotypes which express or lack the InsR. Insulin, at a concentration of 10 nM, significantly increased total neurite length, the length of the longest neurite and the number of branch points of cultured DRG neurons as compared to neurons cultured in control medium or in the presence of 1 μM insulin. In both the control and the insulin exposed cultures, ∼43% of neurons displayed InsR-immunoreactivity. The proportions of transient receptor potential vanilloid type 1 receptor (TRPV1)-immunoreactive (IR), calcitonin gene-related peptide (CGRP)-IR and Bandeiraea simplicifolia isolectin B4 (IB4)-binding neurons amounted to ∼61%, ∼57%, and ∼31% of DRG neurons IR for the InsR. Of the IB4-positive population only neurons expressing the InsR were responsive to insulin. In contrast, TRPV1-IR nociceptive and CGRP-IR peptidergic neurons showed increased tendency for neurite outgrowth which was further enhanced by insulin. However, the responsiveness of DRG neurons expressing the InsR was superior to populations of DRG neurons which lack this receptor. The findings also revealed that besides the expression of the InsR, inherent properties of peptidergic, but not non-peptidergic nociceptive neurons may also significantly contribute to the mechanisms of neurite outgrowth of DRG neurons. These observations suggest distinct regenerative propensity for differing populations of DRG neurons which is significantly affected through insulin receptor signaling.
Collapse
Affiliation(s)
- Bence András Lázár
- Department of Psychiatry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Physiology, University of Szeged, Szeged, Hungary
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - Laura Pálvölgyi
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - Ildikó Dobos
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - István Nagy
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Péter Sántha
- Department of Physiology, University of Szeged, Szeged, Hungary
| |
Collapse
|
77
|
Bishnoi M, Khare P, Brown L, Panchal SK. Transient receptor potential (TRP) channels: a metabolic TR(i)P to obesity prevention and therapy. Obes Rev 2018; 19:1269-1292. [PMID: 29797770 DOI: 10.1111/obr.12703] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/26/2018] [Accepted: 04/11/2018] [Indexed: 12/13/2022]
Abstract
Cellular transport of ions, especially by ion channels, regulates physiological function. The transient receptor potential (TRP) channels, with 30 identified so far, are cation channels with high calcium permeability. These ion channels are present in metabolically active tissues including adipose tissue, liver, gastrointestinal tract, brain (hypothalamus), pancreas and skeletal muscle, which suggests a potential role in metabolic disorders including obesity. TRP channels have potentially important roles in adipogenesis, obesity development and its prevention and therapy because of their physiological properties including calcium permeability, thermosensation and taste perception, involvement in cell metabolic signalling and hormone release. This wide range of actions means that organ-specific actions are unlikely, thus increasing the possibility of adverse effects. Delineation of responses to TRP channels has been limited by the poor selectivity of available agonists and antagonists. Food constituents that can modulate TRP channels are of interest in controlling metabolic status. TRP vanilloid 1 channels modulated by capsaicin have been the most studied, suggesting that this may be the first target for effective pharmacological modulation in obesity. This review shows that most of the TRP channels are potential targets to reduce metabolic disorders through a range of mechanisms.
Collapse
Affiliation(s)
- M Bishnoi
- Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, S.A.S. Nagar (Mohali), Punjab, India.,Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia
| | - P Khare
- Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, S.A.S. Nagar (Mohali), Punjab, India
| | - L Brown
- Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia.,School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | - S K Panchal
- Functional Foods Research Group, Institute for Agriculture and the Environment, University of Southern Queensland, Toowoomba, QLD, Australia
| |
Collapse
|
78
|
Grote CW, Wilson NM, Katz NK, Guilford BL, Ryals JM, Novikova L, Stehno-Bittel L, Wright DE. Deletion of the insulin receptor in sensory neurons increases pancreatic insulin levels. Exp Neurol 2018; 305:97-107. [PMID: 29649429 PMCID: PMC5963702 DOI: 10.1016/j.expneurol.2018.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 11/24/2022]
Abstract
Insulin is known to have neurotrophic properties and loss of insulin support to sensory neurons may contribute to peripheral diabetic neuropathy (PDN). Here, genetically-modified mice were generated in which peripheral sensory neurons lacked the insulin receptor (SNIRKO mice) to determine whether disrupted sensory neuron insulin signaling plays a crucial role in the development of PDN and whether SNIRKO mice develop symptoms of PDN due to reduced insulin neurotrophic support. Our results revealed that SNIRKO mice were euglycemic and never displayed significant changes in a wide range of sensorimotor behaviors, nerve conduction velocity or intraepidermal nerve fiber density. However, SNIRKO mice displayed elevated serum insulin levels, glucose intolerance, and increased insulin content in the islets of Langerhans of the pancreas. These results contribute to the growing idea that sensory innervation of pancreatic islets is key to regulating islet function and that a negative feedback loop of sensory neuron insulin signaling keeps this regulation in balance. Our results suggest that a loss of insulin receptors in sensory neurons does not lead to peripheral nerve dysfunction. The SNIRKO mice will be a powerful tool to investigate sensory neuron insulin signaling and may give a unique insight into the role that sensory neurons play in modifying islet physiology.
Collapse
Affiliation(s)
- Caleb W Grote
- Department of Orthopedic Surgery, University of Kansas Medical Center, United States
| | - Natalie M Wilson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, United States
| | - Natalie K Katz
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, United States
| | - Brianne L Guilford
- Department of Applied Health, Southern Illinois University Edwardsville, United States
| | - Janelle M Ryals
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, United States
| | - Lesya Novikova
- Physical Therapy & Rehabilitation Science, University of Kansas Medical Center, Southern Illinois University Edwardsville, United States
| | - Lisa Stehno-Bittel
- Physical Therapy & Rehabilitation Science, University of Kansas Medical Center, Southern Illinois University Edwardsville, United States
| | - Douglas E Wright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, United States.
| |
Collapse
|
79
|
Lázár BA, Jancsó G, Nagy I, Horváth V, Sántha P. The insulin receptor is differentially expressed in somatic and visceral primary sensory neurons. Cell Tissue Res 2018; 374:243-249. [PMID: 29955950 DOI: 10.1007/s00441-018-2868-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/04/2018] [Indexed: 10/28/2022]
Abstract
Recent studies demonstrated the expression of the insulin receptor (InsR) and its functional interaction with the transient receptor potential vanilloid type 1 receptor (TRPV1) in primary sensory neurons (PSNs). The present study was undertaken to reveal the target-specific expression of the InsR and its co-localization with the TRPV1 in rat PSNs. We assessed the localization of the InsR and its co-localization with the TRPV1 in PSNs retrogradely labelled with biotin-conjugated wheat germ agglutinin injected into the dorsal hind paw skin, the gastrocnemius muscle, the pancreas and the urinary bladder wall. The largest proportions of retrogradely labelled InsR-immunoreactive neurons were identified among PSNs serving the pancreas (~ 54%) and the urinary bladder (~ 53%). The proportions of retrogradely labelled InsR-immunoreactive neurons innervating the dorsal hind paw skin and the gastrocnemius muscle amounted to ~ 22 and ~ 21%. TRPV1-immunoreactive neurons amounted to ~ 63, ~ 62, ~ 67 and ~ 65% of retrogradely labelled cutaneous, muscle, pancreatic and urinary bladder PSNs, respectively. Co-localization of the TRPV1 with the InsR was observed in ~ 16, ~ 15, ~ 29 and ~ 30% of retrogradely labelled cutaneous, muscle, pancreatic and urinary bladder PSNs. These quantitative immunohistochemical data demonstrate a preponderance of InsR-immunoreactivity among PSNs, which innervate visceral targets. The present findings suggest that visceral spinal PSNs are more likely to be exposed to the modulatory effects of insulin on sensory functions, including neurotrophic, nociceptive and inflammatory processes.
Collapse
Affiliation(s)
- Bence András Lázár
- Department of Psychiatry, University of Szeged, Kálvária sugárút 57, Szeged, H-6725, Hungary. .,Department of Physiology, University of Szeged, Szeged, Hungary.
| | - Gábor Jancsó
- Department of Physiology, University of Szeged, Szeged, Hungary
| | - István Nagy
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Viktor Horváth
- First Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Péter Sántha
- Department of Physiology, University of Szeged, Szeged, Hungary
| |
Collapse
|
80
|
Um J, Jung N, Kim D, Choi S, Lee SH, Son Y, Park KS. Substance P preserves pancreatic β-cells in type 1 and type 2 diabetic mice. Biochem Biophys Res Commun 2018; 499:960-966. [PMID: 29626466 DOI: 10.1016/j.bbrc.2018.04.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/09/2023]
Abstract
Preservation of pancreatic β-cells is required for the development of therapies for type 1 and type 2 diabetes (T1D and T2D, respectively). Our previous study demonstrated that substance P (SP) preserves β-cell populations in mice with streptozotocin-induced T1D. Here, we demonstrated that chronic systemic treatment with SP restored the mass of β-cells both in nonobese diabetic (NOD) mice with T1D or db/db mice with T2D. SP delayed the onset of T1D in NOD mice via immune modulation. SP inhibited immune infiltration into islets and the salivary glands of NOD mice. In db/db mice, SP treatment rescued glucose intolerance. Moreover, SP inhibited apoptosis, as well as the activation of pancreatic stellate cells in pancreatic islets of db/db mice. SP downregulated the number of α-smooth muscle actin (α-SMA) expressing cells in db/db pancreatic islets. Cleaved-caspase-3 expression was reduced in islets of SP-treated db/db mice compared to that in the control. Therefore, these results suggested that SP may preserve pancreatic β-cells through immune modulation and protection from the stimulated activation of pancreatic stellate cells and apoptosis in T1D and T2D, respectively.
Collapse
Affiliation(s)
- Jihyun Um
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Nunggum Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Dongjin Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Sanghyuk Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Sang-Ho Lee
- Kyung Hee University Hospital at Gangdong, Seoul 05278, South Korea
| | - Youngsook Son
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Ki-Sook Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, South Korea; East-West Medical Research Institute, Kyung Hee University, Seoul 02447, South Korea; College of Medicine, Kyung Hee University, Seoul 02447, South Korea; Kyung Hee University Medical Center, Seoul 02447, South Korea.
| |
Collapse
|
81
|
Davis EL, Davis AR, Gugala Z, Olmsted-Davis EA. Is heterotopic ossification getting nervous?: The role of the peripheral nervous system in heterotopic ossification. Bone 2018; 109:22-27. [PMID: 28716552 PMCID: PMC5768468 DOI: 10.1016/j.bone.2017.07.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 12/28/2022]
Abstract
Heterotopic ossification (HO), or de novo bone formation in soft tissue, is often observed following traumatic injury. Recent studies suggest that peripheral nerves may play a key functional role in this process. The results supporting a neurological basis for HO are examined in this article. Evidence supports the fact that BMPs released from bone matrix possess the capacity to induce HO. However, the process cannot be recapitulated using recombinant proteins without extremely high doses suggesting other components are required for this process. Study of injuries that increase risk for HO, i.e. amputation, hip replacement, elbow fracture, burn, and CNS injury suggests that a likely candidate is traumatic injury of adjacent peripheral nerves. Recent studies suggest neuroinflammation may play a key functional role, by its ability to open the blood-nerve barrier (BNB). Barrier opening is characterized by a change in permeability and is experimentally assessed by the ability of Evans blue dye to enter the endoneurium of peripheral nerves. A combination of BMP and barrier opening is required to activate bone progenitors in the endoneurial compartment. This process is referred to as "neurogenic HO".
Collapse
Affiliation(s)
- Eleanor L Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, United States
| | - Alan R Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, United States; Department of Pediatrics - Section Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, United States; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, United States
| | - Zbigniew Gugala
- Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Elizabeth A Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX 77030, United States; Department of Pediatrics - Section Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, United States; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
82
|
The Insulin Receptor Is Colocalized With the TRPV1 Nociceptive Ion Channel and Neuropeptides in Pancreatic Spinal and Vagal Primary Sensory Neurons. Pancreas 2018; 47:110-115. [PMID: 29215540 DOI: 10.1097/mpa.0000000000000959] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Recent observations demonstrated the expression of the insulin receptor (InsR) and its functional interaction with the transient receptor potential vanilloid type 1 receptor (TRPV1) in sensory ganglion neurons. Because sensory nerves are implicated in pancreatic inflammatory processes, we studied the colocalization of the InsR with TRPV1 and proinflammatory neuropeptides in spinal and vagal pancreatic afferent neurons. METHODS Immunohistochemistry and quantitative morphometry were used to analyze the expression of TRPV1, InsR, substance P (SP), and calcitonin gene-related peptide (CGRP) in retrogradely labeled pancreatic dorsal root ganglion (DRG) and nodose ganglion (NG) neurons. RESULTS The proportions of retrogradely labeled pancreatic TRPV1-, InsR-, SP-, and CGRP-immunoreactive neurons amounted to 68%, 48%, 33%, and 54% in DRGs and 64%, 49%, 40%, and 25% in the NGs. Of the labeled DRG and NG neurons, 23% and 35% showed both TRPV1 and InsR immunoreactivity. Colocalization of the InsR with SP or CGRP was demonstrated in 14% and 28% of pancreatic DRG and 24% and 8% of pancreatic NG neurons. CONCLUSIONS The present findings provide morphological basis for possible functional interactions among the nociceptive ion channel TRPV1, the InsR, and the proinflammatory neuropeptides SP and CGRP expressed by pancreatic DRG and NG neurons.
Collapse
|
83
|
Christie S, Wittert GA, Li H, Page AJ. Involvement of TRPV1 Channels in Energy Homeostasis. Front Endocrinol (Lausanne) 2018; 9:420. [PMID: 30108548 PMCID: PMC6079260 DOI: 10.3389/fendo.2018.00420] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/04/2018] [Indexed: 12/25/2022] Open
Abstract
The ion channel TRPV1 is involved in a wide range of processes including nociception, thermosensation and, more recently discovered, energy homeostasis. Tightly controlling energy homeostasis is important to maintain a healthy body weight, or to aid in weight loss by expending more energy than energy intake. TRPV1 may be involved in energy homeostasis, both in the control of food intake and energy expenditure. In the periphery, it is possible that TRPV1 can impact on appetite through control of appetite hormone levels or via modulation of gastrointestinal vagal afferent signaling. Further, TRPV1 may increase energy expenditure via heat production. Dietary supplementation with TRPV1 agonists, such as capsaicin, has yielded conflicting results with some studies indicating a reduction in food intake and increase in energy expenditure, and other studies indicating the converse. Nonetheless, it is increasingly apparent that TRPV1 may be dysregulated in obesity and contributing to the development of this disease. The mechanisms behind this dysregulation are currently unknown but interactions with other systems, such as the endocannabinoid systems, could be altered and therefore play a role in this dysregulation. Further, TRPV1 channels appear to be involved in pancreatic insulin secretion. Therefore, given its plausible involvement in regulation of energy and glucose homeostasis and its dysregulation in obesity, TRPV1 may be a target for weight loss therapy and diabetes. However, further research is required too fully elucidate TRPV1s role in these processes. The review provides an overview of current knowledge in this field and potential areas for development.
Collapse
Affiliation(s)
- Stewart Christie
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Gary A. Wittert
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Hui Li
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Amanda J. Page
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- *Correspondence: Amanda J. Page
| |
Collapse
|
84
|
Mori N, Kurata M, Yamazaki H, Matsumura S, Hashimoto T, Kanazawa K, Nadamoto T, Inoue K, Fushiki T. Allyl isothiocyanate increases carbohydrate oxidation through enhancing insulin secretion by TRPV1. Biosci Biotechnol Biochem 2017; 82:698-708. [PMID: 29207921 DOI: 10.1080/09168451.2017.1407234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The transient receptor potential (TRP) V1 is a cation channel belonging to the TRP channel family and it has been reported to be involved in energy metabolism, especially glucose metabolism. While, we have previously shown that intragastric administration of allyl isothiocyanate (AITC) enhanced glucose metabolism via TRPV1, the underlying mechanism has not been elucidated. In this study, we examined the relationship between insulin secretion and the increase in carbohydrate oxidation due to AITC. Intragastric administration of AITC elevated blood insulin levels in mice and AITC directly enhanced insulin secretion from isolated islets. These observations were not reproduced in TRPV1 knockout mice. Furthermore, AITC did not increase carbohydrate oxidation in streptozotocin-treated mice. These results suggest that intragastric administration of AITC could induce insulin secretion from islets via TRPV1 and that enhancement of insulin secretion was related to the increased carbohydrate oxidation due to AITC.
Collapse
Affiliation(s)
- Noriyuki Mori
- a Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Kyoto , Japan.,b Division of Nutrition Sciences, School of Human Culture , University of Shiga Prefecture , Shiga , Japan
| | - Manami Kurata
- a Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Hanae Yamazaki
- c Laboratory of Ajinomoto Integrative Research for Advanced Dieting, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Shigenobu Matsumura
- a Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Takashi Hashimoto
- d Department of Agrobioscience, Graduate School of Agricultural Science , Kobe University , Kobe , Japan
| | - Kazuki Kanazawa
- d Department of Agrobioscience, Graduate School of Agricultural Science , Kobe University , Kobe , Japan
| | - Tomonori Nadamoto
- b Division of Nutrition Sciences, School of Human Culture , University of Shiga Prefecture , Shiga , Japan
| | - Kazuo Inoue
- a Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| | - Tohru Fushiki
- a Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture , Kyoto University , Kyoto , Japan
| |
Collapse
|
85
|
A Chitosan-Based Liposome Formulation Enhances the In Vitro Wound Healing Efficacy of Substance P Neuropeptide. Pharmaceutics 2017; 9:pharmaceutics9040056. [PMID: 29211047 PMCID: PMC5750662 DOI: 10.3390/pharmaceutics9040056] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/09/2017] [Accepted: 12/02/2017] [Indexed: 02/02/2023] Open
Abstract
Currently, there is considerable interest in developing innovative biodegradable nanoformulations for controlled administration of therapeutic proteins and peptides. Substance P (SP) is a neuropeptide of 11 amino acids that belongs to the tachykinins family and it plays an important role in wound healing. However, SP is easily degradable in vivo and has a very short half-life, so the use of chitosan-based nanocarriers could enhance its pharmaceutical properties. In light of the above, the aim of this work was to produce and characterize chitosan-coated liposomes loaded with SP (SP-CH-LP) as novel biomaterials with potential application in mucosal wound healing. The loaded system’s biophysical properties were characterized by dynamic light scattering with non-invasive back scattering (DLS-NIBS), mixed mode measurements and phase analysis light scattering (M3-PALS) and high performance liquid chromatography with ultraviolet/visible light detection (HPLC-UV/VIS). Then, the efficacy of the obtained nanoformulations was examined via proof-of-principle experiments using in vitro cell assays. These assays showed an increment on cell motility and proliferation after treatment with free and encapsulated neuropeptides. Additionally, the effect of SP on wound healing was enhanced by the entrapment on CH-LP. Overall, the amenability of chitosan-based nanomaterials to encapsulate peptides and proteins constitutes a promising approach towards potential novel therapies to treat difficult wounds.
Collapse
|
86
|
TRPV1: A Potential Therapeutic Target in Type 2 Diabetes and Comorbidities? Trends Mol Med 2017; 23:1002-1013. [DOI: 10.1016/j.molmed.2017.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/07/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022]
|
87
|
Overactivity of Liver-Related Neurons in the Paraventricular Nucleus of the Hypothalamus: Electrophysiological Findings in db/db Mice. J Neurosci 2017; 37:11140-11150. [PMID: 29038244 DOI: 10.1523/jneurosci.1706-17.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/11/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022] Open
Abstract
Preautonomic neurons in the paraventricular nucleus (PVN) of the hypothalamus play a large role in the regulation of hepatic functions via the autonomic nervous system. Activation of hepatic sympathetic nerves increases glucose and lipid metabolism and contributes to the elevated hepatic glucose production observed in the type 2 diabetic condition. This augmented sympathetic output could originate from altered activity of liver-related PVN neurons. Remarkably, despite the importance of the brain-liver pathway, the cellular properties of liver-related neurons are not known. In this study, we provide the first evidence of overall activity of liver-related PVN neurons. Liver-related PVN neurons were identified with a retrograde, trans-synaptic, viral tracer in male lean and db/db mice and whole-cell patch-clamp recordings were conducted. In db/db mice, the majority of liver-related PVN neurons fired spontaneously; whereas, in lean mice the majority of liver-related PVN neurons were silent, indicating that liver-related PVN neurons are more active in db/db mice. Persistent, tonic inhibition was identified in liver-related PVN neurons; although, the magnitude of tonic inhibitory control was not different between lean and db/db mice. In addition, our study revealed that the transient receptor potential vanilloid type 1-dependent increase of excitatory neurotransmission was reduced in liver-related PVN neurons of db/db mice. These findings demonstrate plasticity of liver-related PVN neurons and a shift toward excitation in a diabetic mouse model. Our study suggests altered autonomic circuits at the level of the PVN, which can contribute to autonomic dysfunction and dysregulation of neural control of hepatic functions including glucose metabolism.SIGNIFICANCE STATEMENT A growing body of evidence suggests the importance of the autonomic control in the regulation of hepatic metabolism, which plays a major role in the development and progression of type 2 diabetes mellitus. Despite the importance of the brain-liver pathway, the overall activity of liver-related neurons in control and diabetic conditions is not known. This is a significant gap in knowledge, which prevents developing strategies to improve glucose homeostasis via altering the brain-liver pathway. One of the key findings of our study is the overall shift toward excitation in liver-related hypothalamic neurons in the diabetic condition. This overactivity may be one of the underlying mechanisms of elevated sympathetic activity known in metabolically compromised patients and animal models.
Collapse
|
88
|
Udit S, Burton M, Rutkowski JM, Lee S, Bookout AL, Scherer PE, Elmquist JK, Gautron L. Na v1.8 neurons are involved in limiting acute phase responses to dietary fat. Mol Metab 2017; 6:1081-1091. [PMID: 29031710 PMCID: PMC5641637 DOI: 10.1016/j.molmet.2017.07.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 07/19/2017] [Accepted: 07/24/2017] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE AND METHODS Metabolic viscera and their vasculature are richly innervated by peripheral sensory neurons. Here, we examined the metabolic and inflammatory profiles of mice with selective ablation of all Nav1.8-expressing primary afferent neurons. RESULTS While mice lacking sensory neurons displayed no differences in body weight, food intake, energy expenditure, or body composition compared to controls on chow diet, ablated mice developed an exaggerated inflammatory response to high-fat feeding characterized by bouts of weight loss, splenomegaly, elevated circulating interleukin-6 and hepatic serum amyloid A expression. This phenotype appeared to be directly mediated by the ingestion of saturated lipids. CONCLUSIONS These data demonstrate that the Nav1.8-expressing afferent neurons are not essential for energy balance but are required for limiting the acute phase response caused by an obesogenic diet.
Collapse
Affiliation(s)
- Swalpa Udit
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA
| | - Michael Burton
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA
| | - Joseph M Rutkowski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA
| | - Syann Lee
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA
| | - Angie L Bookout
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA; Department of Pharmacology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA
| | - Joel K Elmquist
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA.
| | - Laurent Gautron
- Division of Hypothalamic Research, Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas 75390, TX, USA.
| |
Collapse
|
89
|
Dooley J, Pasciuto E, Lagou V, Lampi Y, Dresselaers T, Himmelreich U, Liston A. NOD mice, susceptible to pancreatic autoimmunity, demonstrate delayed growth of pancreatic cancer. Oncotarget 2017; 8:80167-80174. [PMID: 29113292 PMCID: PMC5655187 DOI: 10.18632/oncotarget.21261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/26/2017] [Indexed: 12/26/2022] Open
Abstract
Pancreatic cancer is a high mortality form of cancer, with a median survival only six months. There are multiple associated risk factors associated, most importantly type 2 diabetes, obesity, pancreatitis and smoking. The relative rarity of the disease, however, has made it difficult to dissect causative risk factors, especially with related risk factors. A major unanswered question with important therapeutic implications is the effect of immunological responses on pancreatic cancer formation, with data from other cancers suggesting the potential for local immunological responses to either increase cancer development or increase cancer elimination. Due to the rarity and late diagnosis of pancreatic cancer direct epidemiological evidence is lacking, thus necessitating a reliance on animal models. Here we investigated the relationship between pancreatic autoimmunity and cancer by backcrossing the well characterised Ela1-Tag transgenic model of pancreatic cancer onto the pancreatic autoimmune susceptible NOD mouse strain. Through longitudinal magnetic resonance imaging we found that the NOD genetic background delayed the onset of pancreatic tumours and substantially slowed the growth rate of tumours after development. These results suggest that elevated autoimmune surveillance of the pancreas limits tumour formation and growth, identifying pancreatic cancer as a promising target for immune checkpoint blockade therapies that unleash latent autoimmunity.
Collapse
Affiliation(s)
- James Dooley
- Translational Immunology Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Emanuela Pasciuto
- Translational Immunology Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Vasiliki Lagou
- Translational Immunology Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Yulia Lampi
- Translational Immunology Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Tom Dresselaers
- Department of Imaging and Pathology, KU Leuven - University of Leuven, Biomedical MRI/MoSAIC, Leuven, Belgium
| | - Uwe Himmelreich
- Department of Imaging and Pathology, KU Leuven - University of Leuven, Biomedical MRI/MoSAIC, Leuven, Belgium
| | - Adrian Liston
- Translational Immunology Laboratory, VIB, Leuven, Belgium.,Department of Microbiology and Immunology, KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
90
|
Pan B, Zhang Z, Chao D, Hogan QH. Dorsal Root Ganglion Field Stimulation Prevents Inflammation and Joint Damage in a Rat Model of Rheumatoid Arthritis. Neuromodulation 2017; 21:247-253. [PMID: 28872725 DOI: 10.1111/ner.12648] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/19/2017] [Accepted: 07/20/2017] [Indexed: 01/30/2023]
Abstract
OBJECTIVES Electrical stimulation of the dorsal root ganglion (DRG), referred to here as ganglionic field stimulation (GFS), is effective in reducing clinical pain, probably by interrupting transmission of afferent impulse trains on sensory neurons as they pass through the DRG. We therefore tested whether efferent impulse trains conveyed by sensory neurons, which contribute to neurogenic inflammation, may also be interrupted by GFS. MATERIALS AND METHODS Collagen-induced arthritis, a model of clinical rheumatoid arthritis, was initiated in rats concurrently with the insertion of an electrode for GFS at the fourth lumbar DRG. Continuous GFS (20 Hz pulse rate, current at 80% of the motor threshold) was initiated 6 days later and continued for 14 days. Plantar pain sensitivity, ankle arthritis score, and dimensions of the foot and ankle were determined one hour after termination of GFS. RESULTS The foot/ankle contralateral to GFS developed hypersensitivity to threshold and noxious mechanical stimulation, swelling, and high arthritis score, all of which were normalized in the foot/ankle ipsilateral with GFS. Histology showed GFS limited joint destruction. Electrophysiological recording showed GFS can block efferent impulse trains. CONCLUSIONS Our findings show that GFS can reduce neurogenic inflammation and the resulting joint damage in a model of rheumatoid arthritis, probably by blocking the transit of impulse trains through the DRG. GFS may have clinical utility in limiting joint destruction in inflammatory arthritis such as rheumatoid arthritis.
Collapse
Affiliation(s)
- Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiyong Zhang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Dongman Chao
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Anesthesiology, Clement J. Zablocki VA Medical Center, Milwaukee, WI, USA
| |
Collapse
|
91
|
Shi Z, Greene WK, Nicholls PK, Hu D, Tirnitz-Parker JEE, Yuan Q, Yin C, Ma B. Immunofluorescent characterization of non-myelinating Schwann cells and their interactions with immune cells in mouse mesenteric lymph node. Eur J Histochem 2017; 61:2827. [PMID: 29046050 PMCID: PMC5572407 DOI: 10.4081/ejh.2017.2827] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/22/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
The central nervous system (CNS) influences the immune system in a general fashion by regulating the systemic concentration of humoral substances, whereas the autonomic nervous system communicates specifically with the immune system according to local interactions. Data concerning the mechanisms of this bidirectional crosstalk of the peripheral nervous system (PNS) and immune system remain limited. To gain a better understanding of local interactions of the PNS and immune system, we have used immunofluorescent staining of glial fibrillary acidic protein (GFAP), coupled with confocal microscopy, to investigate the non-myelinating Schwann cell (NMSC)-immune cell interactions in mouse mesenteric lymph nodes. Our results demonstrate i) the presence of extensive NMSC processes and even of cell bodies in each compartment of the mouse mesenteric lymph node; ii) close associations/interactions of NMSC processes with blood vessels (including high endothelial venules) and the lymphatic vessel/sinus; iii) close contacts/associations of NMSC processes with various subsets of dendritic cells (such as CD4+CD11c+, CD8+CD11c+ dendritic cells), macrophages (F4/80+ and CD11b+ macrophages), and lymphocytes. Our novel findings concerning the distribution of NMSCs and NMSC-immune cell interactions inside the mouse lymph node should help to elucidate the mechanisms through which the PNS affects cellular- and humoral-mediated immune responses or vice versa in health and disease.
Collapse
|
92
|
Kim M, Furuzono T, Yamakuni K, Li Y, Kim YI, Takahashi H, Ohue-Kitano R, Jheng HF, Takahashi N, Kano Y, Yu R, Kishino S, Ogawa J, Uchida K, Yamazaki J, Tominaga M, Kawada T, Goto T. 10-oxo-12( Z)-octadecenoic acid, a linoleic acid metabolite produced by gut lactic acid bacteria, enhances energy metabolism by activation of TRPV1. FASEB J 2017; 31:5036-5048. [PMID: 28754711 DOI: 10.1096/fj.201700151r] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 07/17/2017] [Indexed: 11/11/2022]
Abstract
Gut microbiota can regulate the host energy metabolism; however, the underlying mechanisms that could involve gut microbiota-derived compounds remain to be understood. Therefore, in this study, we investigated the effects of KetoA [10-oxo-12(Z)-octadecenoic acid]-a linoleic acid metabolite produced by gut lactic acid bacteria-on whole-body energy metabolism and found that dietary intake of KetoA could enhance energy expenditure in mice, thereby protecting mice from diet-induced obesity. By using Ca2+ imaging and whole-cell patch-clamp methods, KetoA was noted to potently activate transient receptor potential vanilloid 1 (TRPV1) and enhance noradrenalin turnover in adipose tissues. In addition, KetoA up-regulated genes that are related to brown adipocyte functions, including uncoupling protein 1 (UCP1) in white adipose tissue (WAT), which was later diminished in the presence of a β-adrenoreceptor blocker. By using obese and diabetic model KK-Ay mice, we further show that KetoA intake ameliorated obesity-associated metabolic disorders. In the absence of any observed KetoA-induced antiobesity effect or UCP1 up-regulation in TRPV1-deficient mice, we prove that the antiobesity effect of KetoA was caused by TRPV1 activation-mediated browning in WAT. KetoA produced in the gut could therefore be involved in the regulation of host energy metabolism.-Kim, M., Furuzono, T., Yamakuni, K., Li, Y., Kim, Y.-I., Takahashi, H., Ohue-Kitano, R., Jheng, H.-F., Takahashi, N., Kano, Y., Yu, R., Kishino, S., Ogawa, J., Uchida, K., Yamazaki, J., Tominaga, M., Kawada, T., Goto, T. 10-oxo-12(Z)-octadecenoic acid, a linoleic acid metabolite produced by gut lactic acid bacteria, enhances energy metabolism by activation of TRPV1.
Collapse
Affiliation(s)
- Minji Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tomoya Furuzono
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kanae Yamakuni
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Yongjia Li
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Young-Il Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Haruya Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Huei-Fen Jheng
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Nobuyuki Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Yuriko Kano
- Laboratory of Nutrition Chemistry, Faculty of Home Economics, Kobe Women's University, Kobe, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, South Korea
| | - Shigenobu Kishino
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Jun Ogawa
- Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan.,Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kunitoshi Uchida
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Japan.,Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Jun Yamazaki
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan; .,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| |
Collapse
|
93
|
Regulation of Drosophila hematopoietic sites by Activin-β from active sensory neurons. Nat Commun 2017; 8:15990. [PMID: 28748922 PMCID: PMC5537569 DOI: 10.1038/ncomms15990] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/23/2017] [Indexed: 12/21/2022] Open
Abstract
An outstanding question in animal development, tissue homeostasis and disease is how cell populations adapt to sensory inputs. During Drosophila larval development, hematopoietic sites are in direct contact with sensory neuron clusters of the peripheral nervous system (PNS), and blood cells (hemocytes) require the PNS for their survival and recruitment to these microenvironments, known as Hematopoietic Pockets. Here we report that Activin-β, a TGF-β family ligand, is expressed by sensory neurons of the PNS and regulates the proliferation and adhesion of hemocytes. These hemocyte responses depend on PNS activity, as shown by agonist treatment and transient silencing of sensory neurons. Activin-β has a key role in this regulation, which is apparent from reporter expression and mutant analyses. This mechanism of local sensory neurons controlling blood cell adaptation invites evolutionary parallels with vertebrate hematopoietic progenitors and the independent myeloid system of tissue macrophages, whose regulation by local microenvironments remain undefined.
Collapse
|
94
|
Jung N, Um J, Kim DY, Dubon MJ, Byeon Y, Kim D, Son Y, Park KS. Substance P preserves pancreatic β-cells in streptozotocin-induced type 1 diabetic mice. Biochem Biophys Res Commun 2017; 491:958-965. [PMID: 28754588 DOI: 10.1016/j.bbrc.2017.07.142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/24/2017] [Indexed: 01/03/2023]
Abstract
Preservation of the pancreatic β-cell population is required for the development of therapies for diabetes, which is caused by a decrease in β-cells. Here, we demonstrate the antidiabetic effects of substance P (SP) in type 1 diabetes (T1D) mice induced with streptozotocin. SP enhanced the compensatory proliferation of β-cells in order to restore β-cells in response to acute injury induced by a single high-dose of streptozotocin. However, SP affected neither the basal proliferation of β-cells nor their apoptosis. In vitro studies by using the INS-1 pancreatic β-cell line showed that SP mediated the increase in the proliferation of β-cells via the activation of Akt. Chronic systemic treatment with SP restored the mass of β-cells and inhibited insulitis in T1D mice induced with multiple low-doses of streptozotocin. Therefore, systemic treatment with SP may be a promising therapeutic strategy for treating diabetes in patients with loss of functional β-cells.
Collapse
MESH Headings
- Acute Disease
- Animals
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/prevention & control
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Injections, Intraperitoneal
- Insulin-Secreting Cells/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Pancreatitis/chemically induced
- Pancreatitis/pathology
- Pancreatitis/prevention & control
- Streptozocin/administration & dosage
- Structure-Activity Relationship
- Substance P/pharmacology
Collapse
Affiliation(s)
- Nunggum Jung
- Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Jihyun Um
- Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Do Yeon Kim
- St. Peter's Hospital and R&D Center, Cell & Bio Inc., Seoul 06286, South Korea
| | - Maria Jose Dubon
- Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Yeji Byeon
- Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Dongjin Kim
- Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Youngsook Son
- Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea.
| | - Ki-Sook Park
- East-West Medical Research Institute, Kyung Hee University, Seoul 02447, South Korea; College of Medicine, Kyung Hee University, Seoul 02447, South Korea.
| |
Collapse
|
95
|
Lundberg M, Lindqvist A, Wierup N, Krogvold L, Dahl-Jørgensen K, Skog O. The density of parasympathetic axons is reduced in the exocrine pancreas of individuals recently diagnosed with type 1 diabetes. PLoS One 2017. [PMID: 28628651 PMCID: PMC5476281 DOI: 10.1371/journal.pone.0179911] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
To elucidate the etiology of type 1 diabetes, the affected pancreas needs to be thoroughly characterized. Pancreatic innervation has been suggested to be involved in the pathology of the disease and a reduction of sympathetic innervation of the islets was recently reported. In the present study, we hypothesized that parasympathetic innervation would be altered in the type 1 diabetes pancreas. Human pancreatic specimens were obtained from a unique cohort of individuals with recent onset or long standing type 1 diabetes. Density of parasympathetic axons was assessed by immunofluorescence and morphometry. Our main finding was a reduced density of parasympathetic axons in the exocrine, but not endocrine compartment of the pancreas in individuals with recent onset type 1 diabetes. The reduced density of parasympathetic axons in the exocrine compartment could have functional implications, e.g. be related to the exocrine insufficiency reported in type 1 diabetes patients. Further studies are needed to understand whether reduced parasympathetic innervation is a cause or consequence of type 1 diabetes.
Collapse
Affiliation(s)
- Marcus Lundberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- * E-mail:
| | | | - Nils Wierup
- Lund University Diabetes Centre, Lund University, Malmö, Sweden
| | - Lars Krogvold
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
96
|
Lee Y, Cha YS, Park Y, Lee M. PPARγ2 C1431T Polymorphism Interacts with the Antiobesogenic Effects of Kochujang, a Korean Fermented, Soybean-Based Red Pepper Paste, in Overweight/Obese Subjects: A 12-Week, Double-Blind Randomized Clinical Trial. J Med Food 2017; 20:610-617. [PMID: 28622115 DOI: 10.1089/jmf.2016.3911] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Kochujang, a Korean fermented soybean-based red pepper paste, has been reported to have beneficial health effects. The aim of this study was to examine the antiobesity effects of Kochujang as a supplement in overweight/obese subjects according polymorphisms in the obesity-linked gene, peroxisome proliferator activator receptor γ (PPARγ2). Sixty overweight/obese subjects, who had body mass indexes (BMI, kg/m2) ≥23 or waist/hip ratios (WHR) ≥0.90 for males or ≥0.85 for females, were randomly assigned to either taking 32 g/day of placebo or Kochujang for 12 weeks. Before and after the intervention, anthropometric and metabolic parameters and body fat distribution (by computed tomography) were measured. After PPARγ2 C1431T polymorphism was analyzed by PCR-restriction fragment length polymorphism, the differences among the four groups (wild and mutant alleles in Kochujang and placebo groups) were determined. Between the Kochujang (n = 26) and placebo (n = 27) groups, there were no differences in body composition, insulin resistance, or antioxidant biomarkers before and after intervention. Compared to placebo, Kochujang significantly decreased plasma triglyceride (TG), TG/high-density lipoprotein (HDL), and dietary intakes of protein, sodium, and potassium after age, sex, and BMI were adjusted. The beneficial effects of Kochujang on lowering of TG and TG/HDL were weakened in subjects with the PPARγ2 mutant T allele with increasing subcutaneous fat area. However, the interaction between Kochujang and the PPARγ2 T allele improved insulin sensitivity. The obesogenic variables affected by the T mutant allele of PPARγ2 C1431T SNP were different in overweight/obese subjects in response to Kochujang.
Collapse
Affiliation(s)
- Yunkyoung Lee
- 1 Department of Food Science and Nutrition, Jeju National University , Jeju, Korea
| | - Youn-Soo Cha
- 2 Department of Food Science and Human Nutrition, Obesity Research Center, Chonbuk National University , Jeonju, Korea
| | - Yongsoon Park
- 3 Department of Food and Nutrition, Hanyang University , Seoul, Korea
| | - Myoungsook Lee
- 4 Department of Food and Nutrition, Research Institute of Obesity Sciences, Sungshin Women's University , Seoul, Korea
| |
Collapse
|
97
|
Shirakawa J, De Jesus DF, Kulkarni RN. Exploring inter-organ crosstalk to uncover mechanisms that regulate β-cell function and mass. Eur J Clin Nutr 2017; 71:896-903. [PMID: 28294170 DOI: 10.1038/ejcn.2017.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 02/07/2023]
Abstract
Impaired β-cell function and insufficient β-cell mass compensation are twin pathogenic features that underlie type 2 diabetes (T2D). Current therapeutic strategies continue to evolve to improve treatment outcomes in different ethnic populations and include approaches to counter insulin resistance and improve β-cell function. Although the effects of insulin secretion on metabolic organs such as liver, skeletal muscle and adipose is directly relevant for improving glucose uptake and reduce hyperglycemia, the ability of pancreatic β-cells to crosstalk with multiple non-metabolic tissues is providing novel insights into potential opportunities for improving β-cell function and/or mass that could have beneficial effects in patients with diabetes. For example, the role of the gastrointestinal system in the regulation of β-cell biology is well recognized and has been exploited clinically to develop incretin-related antidiabetic agents. The microbiome and the immune system are emerging as important players in regulating β-cell function and mass. The rich innervation of islet cells indicates it is a prime organ for regulation by the nervous system. In this review, we discuss the potential implications of signals from these organ systems as well as those from bone, placenta, kidney, thyroid, endothelial cells, reproductive organs and adrenal and pituitary glands that can directly impact β-cell biology. An added layer of complexity is the limited data regarding the relative relevance of one or more of these systems in different ethnic populations. It is evident that better understanding of this paradigm would provide clues to enhance β-cell function and/or mass in vivo in the long-term goal of treating or curing patients with diabetes.
Collapse
Affiliation(s)
- J Shirakawa
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - D F De Jesus
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA.,Graduate Program in Areas of Basic and Applied Biology (GABBA), Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - R N Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
98
|
Agonist-dependence of functional properties for common nonsynonymous variants of human transient receptor potential vanilloid 1. Pain 2017; 157:1515-1524. [PMID: 26967694 DOI: 10.1097/j.pain.0000000000000556] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a polymodal receptor activated by capsaicin, heat, and acid, which plays critical roles in thermosensation and pain. In addition, TRPV1 also contributes to multiple pathophysiological states in respiratory, cardiovascular, metabolic, and renal systems. These contributions are further supported by evidence that variations in the human TRPV1 (hTRPV1) gene are associated with various physiological and pathological phenotypes. However, it is not well understood how the variations in hTRPV1 affect channel functions. In this study, we examined functional consequences of amino acid variations of hTRPV1 induced by 5 nonsynonymous single-nucleotide polymorphisms (SNPs) that most commonly exist in the human population. Using electrophysiological assays in HEK293 cells, we examined 9 parameters: activation, Ca permeation, and desensitization after activation by capsaicin, acid, and heat. Our results demonstrated that the 5 SNPs differentially affected functional properties of hTRPV1 in an agonist-dependent manner. Based upon the directionality of change of each phenotype and cumulative changes in each SNP, we classified the 5 SNPs into 3 presumptive functional categories: gain of function (hTRPV1 Q85R, P91S, and T469I), loss of function (I585V), and mixed (M315I). These results reveal a spectrum of functional variation among common hTRPV1 polymorphisms in humans and may aid mechanistic interpretation of phenotypes associated with nonsynonymous hTRPV1 SNPs under pathophysiological conditions.
Collapse
|
99
|
Marche P, Dubois S, Abraham P, Parot-Schinkel E, Gascoin L, Humeau-Heurtier A, Ducluzeau PH, Mahe G. Neurovascular microcirculatory vasodilation mediated by C-fibers and Transient receptor potential vanilloid-type-1 channels (TRPV 1) is impaired in type 1 diabetes. Sci Rep 2017; 7:44322. [PMID: 28287157 PMCID: PMC5347083 DOI: 10.1038/srep44322] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/06/2017] [Indexed: 01/20/2023] Open
Abstract
Microvascular dysfunction may have an early onset in type 1 diabetes (T1D) and can precede major complications. Our objectives were to assess the endothelial-dependent (acetylcholine, ACh; and post-occlusive hyperemia, PORH), non-endothelial-dependent (sodium nitroprusside, SNP) and neurovascular-dependent (local heating, LH and current induced vasodilation, CIV) microcirculatory vasodilation in T1D patients compared with matched control subjects using a laser speckle contrast imager. Seventeen T1D patients - matched with 17 subjects according to age, gender, Body-Mass-Index, and smoking status - underwent macro- and microvascular investigations. The LH early peak assessed the transient receptor potential vanilloid type 1 channels (TRPV1) mediated vasodilation, whereas the plateau assessed the Nitirc-Oxyde (NO) and endothelium-derived hyperpolarizing factor (EDHF) pathways. PORH explored sensory nerves and (EDHF), while CIV assessed sensory nerves (C-fibers) and prostaglandin-mediated vasodilation. Using neurological investigations, we observed that C-fiber and A-delta fiber functions in T1D patients were similar to control subjects. PORH, CIV, LH peak and plateau vasodilations were significantly decreased in T1D patients compared to controls, whereas there was no difference between the two groups for ACh and SNP vasodilations. Neurovascular microcirculatory vasodilations (C-fibers and TRPV 1-mediated vasodilations) are impaired in TD1 patients whereas no abnormalities were found using clinical neurological investigations. Clinicaltrials: No. NCT02538120.
Collapse
Affiliation(s)
- P Marche
- Endocrinology Department, University Hospital of Angers, Angers, France
| | - S Dubois
- Endocrinology Department, University Hospital of Angers, Angers, France
| | - P Abraham
- Laboratory of Vascular Investigations, University Hospital of Angers, UMR CNRS6214/INSERM1083, LUNAM Université, Angers, France
| | - E Parot-Schinkel
- Methodology and Biostatistics Unit, University Hospital of Angers, Angers, France
| | - L Gascoin
- Laboratory of Vascular Investigations, University Hospital of Angers, UMR CNRS6214/INSERM1083, LUNAM Université, Angers, France
| | - A Humeau-Heurtier
- University of Angers, LARIS - Laboratoire Angevin de Recherche en Ingénierie des Systèmes, Angers, France
| | - P H Ducluzeau
- Endocrinology Department, University Hospital of Tours, Tours, France
| | - G Mahe
- INSERM Clinical Investigation Center (CIC 14 14), Rennes, France, Université de Rennes 1 and LUNAM University, Inserm 1083/CNRS 6214, Faculty of Medicine, Angers, France
| |
Collapse
|
100
|
Bougnères P, Le Fur S, Valtat S, Kamatani Y, Lathrop M, Valleron AJ. Using spatio-temporal surveillance data to test the infectious environment of children before type 1 diabetes diagnosis. PLoS One 2017; 12:e0170658. [PMID: 28152013 PMCID: PMC5289461 DOI: 10.1371/journal.pone.0170658] [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: 05/19/2016] [Accepted: 01/09/2017] [Indexed: 12/16/2022] Open
Abstract
The "hygiene hypothesis" postulates that reduced exposure to infections favours the development of autoimmunity and childhood type 1 diabetes (T1D). But on the other side, viruses, notably enteroviruses, are suspected to trigger T1D. The assessment of the possible relationships between infections and T1D still defies the classical tools of epidemiology. We report the methods and results of a geographical approach that maps the addresses of patients to a communicable diseases surveillance database. We mapped the addresses of patients at birth, infancy and T1D diagnosis to the weekly estimates of the regional incidences of 5 frequent communicable diseases routinely collected since 1984 by the French Sentinel network. The pre-diagnostic infectious environment of 3548 patients with T1D diagnosed between 0.5 and 15 years was compared to those of 100 series of age-matched "virtual controls" drawn randomly on the map. Associations were classified as "suggestive" (summer diarrhea, SD, and varicella, V) when p< 0.05, or "significant" (influenza-like infections, ILI) when they passed the Bonferroni correction for FDR. Exposure to ILI and SD were associated with T1D risk, while V seemed protective. In the subset of 2521 patients for which we had genome wide data, we used a case-only approach to search for interactions between SNPs and the infectious environment as defined by the Sentinel database. Two SNPs, rs116624278 and rs77232854, showed significant interaction with exposure to V between 1 and 3 years of life. The infectious associations found should be taken as possible markers of patients' environment, not as direct causative factors of T1D. They require replication in other populations. The increasing public availability of geographical environmental databases will expand the present approach to map thousands of environmental factors to the lifeline of patients affected by various diseases.
Collapse
Affiliation(s)
- Pierre Bougnères
- Department of Pediatric Endocrinology, Bicêtre Hospital, Paris Sud University, AP-HP, Le Kremlin Bicêtre, France
- Inserm U1169, Paris Sud University, Le Kremlin Bicêtre, France
- * E-mail:
| | - Sophie Le Fur
- Department of Pediatric Endocrinology, Bicêtre Hospital, Paris Sud University, AP-HP, Le Kremlin Bicêtre, France
- Inserm U1169, Paris Sud University, Le Kremlin Bicêtre, France
| | | | - Sophie Valtat
- Department of Pediatric Endocrinology, Bicêtre Hospital, Paris Sud University, AP-HP, Le Kremlin Bicêtre, France
| | - Yoichiro Kamatani
- Center for Integrative Medical Sciences, RIKEN, Laboratory for Statistical Analysis, Kanagawa, Japan
| | - Mark Lathrop
- Centre National de Génotypage, Evry, France, and Génome Québec Innovation Centre, McGill University, Montréal (Québec), Canada
| | | |
Collapse
|