1
|
Tahiri A, Youssef A, Inoue R, Moon S, Alsarkhi L, Berroug L, Nguyen XTA, Wang L, Kwon H, Pang ZP, Zhao JY, Shirakawa J, Ulloa L, El Ouaamari A. Vagal sensory neuron-derived FGF3 controls insulin secretion. Dev Cell 2025; 60:51-61.e4. [PMID: 39413782 PMCID: PMC11706709 DOI: 10.1016/j.devcel.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/28/2023] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
Vagal nerve stimulation has emerged as a promising modality for treating a wide range of chronic conditions, including metabolic disorders. However, the cellular and molecular pathways driving these clinical benefits remain largely obscure. Here, we demonstrate that fibroblast growth factor 3 (Fgf3) mRNA is upregulated in the mouse vagal ganglia under acute metabolic stress. Systemic and vagal sensory overexpression of Fgf3 enhanced glucose-stimulated insulin secretion (GSIS), improved glucose excursion, and increased energy expenditure and physical activity. Fgf3-elicited insulinotropic and glucose-lowering responses were recapitulated when overexpression of Fgf3 was restricted to the pancreas-projecting vagal sensory neurons. Genetic ablation of Fgf3 in pancreatic vagal afferents exacerbated high-fat diet-induced glucose intolerance and blunted GSIS. Finally, electrostimulation of the vagal afferents enhanced GSIS and glucose clearance independently of efferent outputs. Collectively, we demonstrate a direct role for the vagal afferent signaling in GSIS and identify Fgf3 as a vagal sensory-derived metabolic factor that controls pancreatic β-cell activity.
Collapse
Affiliation(s)
- Azeddine Tahiri
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 01595, USA
| | - Ayman Youssef
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC 27710, USA
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Sohyun Moon
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Lamyaa Alsarkhi
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 01595, USA
| | - Laila Berroug
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 01595, USA
| | - Xuan Thi Anh Nguyen
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Le Wang
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Hyokjoon Kwon
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Jerry Yingtao Zhao
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Luis Ulloa
- Center for Perioperative Organ Protection, Department of Anesthesiology, Duke University, Durham, NC 27710, USA
| | - Abdelfattah El Ouaamari
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 01595, USA; Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA.
| |
Collapse
|
2
|
Shen YR, Cheng L, Zhang DF. TRPV1: A novel target for the therapy of diabetes and diabetic complications. Eur J Pharmacol 2024; 984:177021. [PMID: 39362389 DOI: 10.1016/j.ejphar.2024.177021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Diabetes mellitus is a chronic metabolic disease characterized by abnormally elevated blood glucose levels. Type II diabetes accounts for approximately 90% of all cases. Several drugs are available for hyperglycemia treatment. However, the current therapies for managing high blood glucose do not prevent or reverse the disease progression, which may result in complications and adverse effects, including diabetic neuropathy, retinopathy, and nephropathy. Hence, developing safer and more effective methods for lowering blood glucose levels is imperative. Transient receptor potential vanilloid-1 (TRPV1) is a significant member of the transient receptor potential family. It is present in numerous body tissues and organs and performs vital physiological functions. PURPOSE This review aimed to develop new targeted TRPV1 hypoglycemic drugs by systematically summarizing the mechanism of action of the TRPV1-based signaling pathway in preventing and treating diabetes and its complications. METHODS Literature searches were performed in the PubMed, Web of Science, Google Scholar, Medline, and Scopus databases for 10 years from 2013 to 2023. The search terms included "diabetes," "TRPV1," "diabetic complications," and "capsaicin." RESULTS TRPV1 is an essential potential target for treating diabetes mellitus and its complications. It reduces hepatic glucose production and food intake and promotes thermogenesis, metabolism, and insulin secretion. Activation of TRPV1 ameliorates diabetic nephropathy, retinopathy, myocardial infarction, vascular endothelial dysfunction, gastroparesis, and bladder dysfunction. Suppression of TRPV1 improves diabetes-related osteoporosis. However, the therapeutic effects of activating or suppressing TRPV1 may vary when treating diabetic neuropathy and periodontitis. CONCLUSION This review demonstrates that TRPV1 is a potential therapeutic target for diabetes and its complications. Additionally, it provides a theoretical basis for developing new hypoglycemic drugs that target TRPV1.
Collapse
Affiliation(s)
- Yu-Rong Shen
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Long Cheng
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Dong-Fang Zhang
- Department of Pharmacognosy, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
3
|
Mikhailov N, Hämäläinen RH. Modulating Mitochondrial DNA Heteroplasmy with Mitochondrially Targeted Endonucleases. Ann Biomed Eng 2024; 52:2627-2640. [PMID: 36001180 PMCID: PMC11329604 DOI: 10.1007/s10439-022-03051-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/09/2022] [Indexed: 11/01/2022]
Abstract
Mitochondria, mainly known as energy factories of eukaryotic cells, also exert several additional signaling and metabolic functions and are today recognized as major cellular biosynthetic and signaling hubs. Mitochondria possess their own genome (mitochondrial DNA-mtDNA), that encodes proteins essential for oxidative phosphorylation, and mutations in it are an important contributor to human disease. The mtDNA mutations often exist in heteroplasmic conditions, with both healthy and mutant versions of the mtDNA residing in patients' cells and the level of mutant mtDNA may vary between different tissues and organs and affect the clinical outcome of the disease. Thus, shifting the ratio between healthy and mutant mtDNA in patients' cells provides an intriguing therapeutic option for mtDNA diseases. In this review we describe current strategies for modulating mitochondrial heteroplasmy levels with engineered endonucleases including mitochondrially targeted TALENs and Zinc finger nucleases (ZFNs) and discuss their therapeutic potential. These gene therapy tools could in the future provide therapeutic help both for patients with mitochondrial disease as well as in preventing the transfer of pathogenic mtDNA mutations from a mother to her offspring.
Collapse
Affiliation(s)
- Nikita Mikhailov
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland
| | - Riikka H Hämäläinen
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211, Kuopio, Finland.
| |
Collapse
|
4
|
Howe CL, Icka-Araki D, Viray AEG, Garza S, Frank JA. Optical Control of TRPV1 Channels In Vitro with Tethered Photopharmacology. ACS Chem Biol 2024; 19:1466-1473. [PMID: 38904446 DOI: 10.1021/acschembio.4c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a nonselective cation channel that is important for nociception and inflammatory pain and is activated by a variety of nociceptive stimuli─including lipids such as capsaicin (CAP) and endocannabinoids. TRPV1's role in physiological systems is often studied by activating it with externally perfused ligands; however, this approach is plagued by poor spatiotemporal resolution. Lipid agonists are insoluble in physiological buffers and can permeate membranes to accumulate nonselectively inside cells, where they can have off-target effects. To increase the spatiotemporal precision with which we can activate lipids on cells and tissues, we previously developed optically cleavable targeted (OCT) ligands, which use protein tags (SNAP-tags) to localize a photocaged ligand on a target cellular membrane. After enrichment, the active ligand is released on a flash of light to activate nearby receptors. In our previous work, we developed an OCT-ligand to control a cannabinoid-sensitive GPCR. Here, we expand the scope of OCT-ligand technology to target TRPV1 ion channels. We synthesize a probe, OCT-CAP, that tethers to membrane-bound SNAP-tags and releases a TRPV1 agonist when triggered by UV-A irradiation. Using Ca2+ imaging and electrophysiology in HEK293T cells expressing TRPV1, we demonstrate that OCT-CAP uncaging activates TRPV1 with superior spatiotemporal precision when compared to standard diffusible ligands or photocages. This study is the first example of an OCT-ligand designed to manipulate an ion-channel target. We anticipate that these tools will find many applications in controlling lipid signaling pathways in various cells and tissues.
Collapse
Affiliation(s)
- Carmel L Howe
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - David Icka-Araki
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - Alexander E G Viray
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - Sarahi Garza
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239, United States
- Neuroscience Graduate Program, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - James A Frank
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon 97239, United States
- Vollum Institute, Oregon Health & Science University, Portland, Oregon 97239, United States
| |
Collapse
|
5
|
Liu C, Wang W, Zhao S, Chen S, Chen H, Wang S, Li Z, Qian H, Tian X. Discovery of first-in-class highly selective TRPV1 antagonists with dual analgesic and hypoglycemic effects. Bioorg Med Chem 2024; 107:117750. [PMID: 38776567 DOI: 10.1016/j.bmc.2024.117750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
Analgesia and blood sugar control are considered as two main unmet clinical needs for diabetes related neuropathic pain patients. Transient receptor potential vanilloid type-1 (TRPV1) channel is a highly validated target for pain perception, while no TRPV1 antagonists have been approved due to hyperthermia side effects. Herein, two series of new TRPV1 antagonists with flavonoid skeleton were designed by the structure-based drug design (SBDD) strategy. After comprehensive evaluation, compound CX-3 was identified as a promising TRPV1 antagonist. CX-3 exhibited equivalent TRPV1 antagonistic activity with classical TRPV1 antagonist BCTC in vitro, and exerted better analgesic activity in vivo than that of BCTC in the formalin induced inflammatory pain model without hyperthermia risk. Moreover, CX-3 exhibited robust glucose-lowering effects and showed high selectivity over other ion channels. Overall, these findings identified a first-in-class highly selective TRPV1 antagonist CX-3, which is a promising candidate to target the pathogenesis of diabetes related neuropathic pain.
Collapse
Affiliation(s)
- Chunxia Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Wenxin Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Shiyu Zhao
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siliang Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Haoyang Chen
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Suhua Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China.
| | - Xin Tian
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Henan Key Laboratory of Precision Clinical Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
6
|
Tynan A, Tsaava T, Gunasekaran M, Bravo Iñiguez CE, Brines M, Chavan SS, Tracey KJ. TRPV1 nociceptors are required to optimize antigen-specific primary antibody responses to novel antigens. Bioelectron Med 2024; 10:14. [PMID: 38807193 PMCID: PMC11134756 DOI: 10.1186/s42234-024-00145-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Key to the advancement of the field of bioelectronic medicine is the identification of novel pathways of neural regulation of immune function. Sensory neurons (termed nociceptors) recognize harmful stimuli and initiate a protective response by eliciting pain and defensive behavior. Nociceptors also interact with immune cells to regulate host defense and inflammatory responses. However, it is still unclear whether nociceptors participate in regulating primary IgG antibody responses to novel antigens. METHODS To understand the role of transient receptor potential vanilloid 1 (TRPV1)-expressing neurons in IgG responses, we generated TRPV1-Cre/Rosa-ChannelRhodopsin2 mice for precise optogenetic activation of TRPV1 + neurons and TRPV1-Cre/Lox-diphtheria toxin A mice for targeted ablation of TRPV1-expressing neurons. Antigen-specific antibody responses were longitudinally monitored for 28 days. RESULTS Here we show that TRPV1 expressing neurons are required to develop an antigen-specific immune response. We demonstrate that selective optogenetic stimulation of TRPV1+ nociceptors during immunization significantly enhances primary IgG antibody responses to novel antigens. Further, mice rendered deficient in TRPV1- expressing nociceptors fail to develop primary IgG antibody responses to keyhole limpet hemocyanin or haptenated antigen. CONCLUSION This functional and genetic evidence indicates a critical role for nociceptor TRPV1 in antigen-specific primary antibody responses to novel antigens. These results also support consideration of potential therapeutic manipulation of nociceptor pathways using bioelectronic devices to enhance immune responses to foreign antigens.
Collapse
Affiliation(s)
- Aisling Tynan
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Téa Tsaava
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Manojkumar Gunasekaran
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Carlos E Bravo Iñiguez
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA
| | - Michael Brines
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sangeeta S Chavan
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hofstra University, Hempstead, NY, USA.
| | - Kevin J Tracey
- Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA.
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hofstra University, Hempstead, NY, USA.
| |
Collapse
|
7
|
Maximiano TKE, Carneiro JA, Fattori V, Verri WA. TRPV1: Receptor structure, activation, modulation and role in neuro-immune interactions and pain. Cell Calcium 2024; 119:102870. [PMID: 38531262 DOI: 10.1016/j.ceca.2024.102870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024]
Abstract
In the 1990s, the identification of a non-selective ion channel, especially responsive to capsaicin, revolutionized the studies of somatosensation and pain that were to follow. The TRPV1 channel is expressed mainly in neuronal cells, more specifically, in sensory neurons responsible for the perception of noxious stimuli. However, its presence has also been detected in other non-neuronal cells, such as immune cells, β- pancreatic cells, muscle cells and adipocytes. Activation of the channel occurs in response to a wide range of stimuli, such as noxious heat, low pH, gasses, toxins, endocannabinoids, lipid-derived endovanilloid, and chemical agents, such as capsaicin and resiniferatoxin. This activation results in an influx of cations through the channel pore, especially calcium. Intracellular calcium triggers different responses in sensory neurons. Dephosphorylation of the TRPV1 channel leads to its desensitization, which disrupts its function, while its phosphorylation increases the channel's sensitization and contributes to the channel's rehabilitation after desensitization. Kinases, phosphoinositides, and calmodulin are the main signaling pathways responsible for the channel's regulation. Thus, in this review we provide an overview of TRPV1 discovery, its tissue expression as well as on the mechanisms by which TRPV1 activation (directly or indirectly) induces pain in different disease models.
Collapse
Affiliation(s)
- Thaila Kawane Euflazio Maximiano
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Jessica Aparecida Carneiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil
| | - Victor Fattori
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital-Harvard Medical School, Karp Research Building, 300 Longwood Ave, 02115, Boston, Massachusetts, United States.
| | - Waldiceu A Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina, Paraná, Brazil.
| |
Collapse
|
8
|
Yang B, Ma D, Zhu X, Wu Z, An Q, Zhao J, Gao X, Zhang L. Roles of TRP and PIEZO receptors in autoimmune diseases. Expert Rev Mol Med 2024; 26:e10. [PMID: 38659380 PMCID: PMC11140548 DOI: 10.1017/erm.2023.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/15/2023] [Accepted: 08/21/2023] [Indexed: 04/26/2024]
Abstract
Autoimmune diseases are pathological autoimmune reactions in the body caused by various factors, which can lead to tissue damage and organ dysfunction. They can be divided into organ-specific and systemic autoimmune diseases. These diseases usually involve various body systems, including the blood, muscles, bones, joints and soft tissues. The transient receptor potential (TRP) and PIEZO receptors, which resulted in David Julius and Ardem Patapoutian winning the Nobel Prize in Physiology or Medicine in 2021, attracted people's attention. Most current studies on TRP and PIEZO receptors in autoimmune diseases have been carried out on animal model, only few clinical studies have been conducted. Therefore, this study aimed to review existing studies on TRP and PIEZO to understand the roles of these receptors in autoimmune diseases, which may help elucidate novel treatment strategies.
Collapse
Affiliation(s)
- Baoqi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xueqing Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Zewen Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| |
Collapse
|
9
|
Borgmann D, Fenselau H. Vagal pathways for systemic regulation of glucose metabolism. Semin Cell Dev Biol 2024; 156:244-252. [PMID: 37500301 DOI: 10.1016/j.semcdb.2023.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 06/20/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Maintaining blood glucose at an appropriate physiological level requires precise coordination of multiple organs and tissues. The vagus nerve bidirectionally connects the central nervous system with peripheral organs crucial to glucose mobilization, nutrient storage, and food absorption, thereby presenting a key pathway for the central control of blood glucose levels. However, the precise mechanisms by which vagal populations that target discrete tissues participate in glucoregulation are much less clear. Here we review recent advances unraveling the cellular identity, neuroanatomical organization, and functional contributions of both vagal efferents and vagal afferents in the control of systemic glucose metabolism. We focus on their involvement in relaying glucoregulatory cues from the brain to peripheral tissues, particularly the pancreatic islet, and by sensing and transmitting incoming signals from ingested food to the brain. These recent findings - largely driven by advances in viral approaches, RNA sequencing, and cell-type selective manipulations and tracings - have begun to clarify the precise vagal neuron populations involved in the central coordination of glucose levels, and raise interesting new possibilities for the treatment of glucose metabolism disorders such as diabetes.
Collapse
Affiliation(s)
- Diba Borgmann
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Physical Activity Research (CFAS), Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Henning Fenselau
- Synaptic Transmission in Energy Homeostasis Group, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Strasse 26, 50937 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, Cologne 50931, Germany.
| |
Collapse
|
10
|
Ding X, Chen J, Zeng W. Neuroimmune regulation in the pancreas. FUNDAMENTAL RESEARCH 2024; 4:201-205. [PMID: 38933519 PMCID: PMC11197567 DOI: 10.1016/j.fmre.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/13/2022] [Accepted: 08/01/2022] [Indexed: 11/21/2022] Open
Abstract
The pancreas exerts endocrine and exocrine functions in energy balance. The neural innervation and immune milieu are both crucial in supporting pancreatic homeostasis. The neuronal network connects the pancreas with the central nervous system (CNS) and the enteric nervous system (ENS) and sustains metabolic activities. The nerves in the pancreas are categorized as spinal sensory afferent fibers, vagal sensory afferent nerves, autonomic fibers of both sympathetic and parasympathetic divisions, and fibers from the ENS and intrapancreatic ganglia. They innervate different regions and various cell types, which collectively determine physiological functions. Studies have established that the diverse pathological conditions, including pancreatitis, diabetes, and pancreatic tumor, are attributed to aberrant immune reactions; however, it is largely not clear how the neuronal network may influence the disease conditions. Enlightened by the recent advances illuminating the organ-wide neuronal architecture and the dysfunctions in pancreatic disorders, this review will highlight emerging opportunities to explore the cellular interrelationship, particularly the neuroimmune components in pancreatic health and diseases.
Collapse
Affiliation(s)
- Xiaofan Ding
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jianhui Chen
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Wenwen Zeng
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
11
|
Christoffersson G, Fousteri G. Editorial: Footprints of immune cells in the type 1 diabetic pancreas, volume II. Front Endocrinol (Lausanne) 2024; 15:1367245. [PMID: 38379865 PMCID: PMC10877058 DOI: 10.3389/fendo.2024.1367245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/29/2024] [Indexed: 02/22/2024] Open
Affiliation(s)
| | - Georgia Fousteri
- Division of Immunology, Transplantation, and Infectious Diseases, Diabetes Research Institute, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Hospital, Milan, Italy
| |
Collapse
|
12
|
Zeng J, Lu Y, Chu H, Lu L, Chen Y, Ji K, Lin Y, Li J, Wang S. Research trends and frontier hotspots of TRPV1 based on bibliometric and visualization analyses. Heliyon 2024; 10:e24153. [PMID: 38293347 PMCID: PMC10827456 DOI: 10.1016/j.heliyon.2024.e24153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
Background Transient receptor potential vanilloid type1 (TRPV1) is a non-selective cation channel with multiple activation mechanisms, which has received increasing attention since it was first cloned in 1997. Methods We used bibliometric and visualization analyses to evaluate the theme trends and knowledge structure of TRPV1 research-papers on TRPV1 from 2002 to 2022 obtained from the Web of Science Core Collection. VOSviewer and CiteSpace were used to analyze authors, institutions, countries, co-cited references, and keywords. Results A total of 7413 papers were included. The main research area of TRPV1 was neuroscience; the most published country was the United States, and the University of California, San Francisco, had the highest centrality. Two major collaborative sub-networks were formed between the authors. The distribution of keywords shows that TRPV1 was initially studied extensively, and the recent studies focused on TRPV1 structure and diseases. "Oxidative stress," "TRPV1 structure," "cancer," and "model" have been the research hotspots in recent years. Conclusions This research provides valuable information for the study of TRPV1. Disease research was focused on pain, cancer, and neurodegenerative diseases. Both agonists and antagonists of TRPV1 are gradually being used in clinical practice, and acupuncture was effective in treating TRPV1-mediated inflammatory pain. TRPV1 is involved in classical endogenous cannabis system signaling, and new signaling pathways continue to be revealed.
Collapse
Affiliation(s)
- Jingchun Zeng
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yiqian Lu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hui Chu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liming Lu
- Clinical Research and Data Center, South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yuexuan Chen
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Kaisong Ji
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yeze Lin
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jingjing Li
- Bao'an Traditional Chinese Medicine Hospital//Seventh Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Shuxin Wang
- Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Amaya-Rodriguez CA, Carvajal-Zamorano K, Bustos D, Alegría-Arcos M, Castillo K. A journey from molecule to physiology and in silico tools for drug discovery targeting the transient receptor potential vanilloid type 1 (TRPV1) channel. Front Pharmacol 2024; 14:1251061. [PMID: 38328578 PMCID: PMC10847257 DOI: 10.3389/fphar.2023.1251061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/14/2023] [Indexed: 02/09/2024] Open
Abstract
The heat and capsaicin receptor TRPV1 channel is widely expressed in nerve terminals of dorsal root ganglia (DRGs) and trigeminal ganglia innervating the body and face, respectively, as well as in other tissues and organs including central nervous system. The TRPV1 channel is a versatile receptor that detects harmful heat, pain, and various internal and external ligands. Hence, it operates as a polymodal sensory channel. Many pathological conditions including neuroinflammation, cancer, psychiatric disorders, and pathological pain, are linked to the abnormal functioning of the TRPV1 in peripheral tissues. Intense biomedical research is underway to discover compounds that can modulate the channel and provide pain relief. The molecular mechanisms underlying temperature sensing remain largely unknown, although they are closely linked to pain transduction. Prolonged exposure to capsaicin generates analgesia, hence numerous capsaicin analogs have been developed to discover efficient analgesics for pain relief. The emergence of in silico tools offered significant techniques for molecular modeling and machine learning algorithms to indentify druggable sites in the channel and for repositioning of current drugs aimed at TRPV1. Here we recapitulate the physiological and pathophysiological functions of the TRPV1 channel, including structural models obtained through cryo-EM, pharmacological compounds tested on TRPV1, and the in silico tools for drug discovery and repositioning.
Collapse
Affiliation(s)
- Cesar A. Amaya-Rodriguez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Departamento de Fisiología y Comportamiento Animal, Facultad de Ciencias Naturales, Exactas y Tecnología, Universidad de Panamá, Ciudad de Panamá, Panamá
| | - Karina Carvajal-Zamorano
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniel Bustos
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
- Laboratorio de Bioinformática y Química Computacional, Departamento de Medicina Traslacional, Facultad de Medicina, Universidad Católica del Maule, Talca, Chile
| | - Melissa Alegría-Arcos
- Núcleo de Investigación en Data Science, Facultad de Ingeniería y Negocios, Universidad de las Américas, Santiago, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado Universidad Católica del Maule, Talca, Chile
| |
Collapse
|
14
|
Agerskov RH, Nyeng P. Innervation of the pancreas in development and disease. Development 2024; 151:dev202254. [PMID: 38265192 DOI: 10.1242/dev.202254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
The autonomic nervous system innervates the pancreas by sympathetic, parasympathetic and sensory branches during early organogenesis, starting with neural crest cell invasion and formation of an intrinsic neuronal network. Several studies have demonstrated that signals from pancreatic neural crest cells direct pancreatic endocrinogenesis. Likewise, autonomic neurons have been shown to regulate pancreatic islet formation, and have also been implicated in type I diabetes. Here, we provide an overview of recent progress in mapping pancreatic innervation and understanding the interactions between pancreatic neurons, epithelial morphogenesis and cell differentiation. Finally, we discuss pancreas innervation as a factor in the development of diabetes.
Collapse
Affiliation(s)
- Rikke Hoegsberg Agerskov
- Roskilde University, Department of Science and Environment, Universitetsvej 1, building 28, Roskilde 4000, Denmark
| | - Pia Nyeng
- Roskilde University, Department of Science and Environment, Universitetsvej 1, building 28, Roskilde 4000, Denmark
| |
Collapse
|
15
|
Vyakaranam AR, Mahamed MM, Hellman P, Eriksson O, Espes D, Christoffersson G, Sundin A. Non-invasive imaging of sympathetic innervation of the pancreas in individuals with type 2 diabetes. Diabetologia 2024; 67:199-208. [PMID: 37935826 PMCID: PMC10709256 DOI: 10.1007/s00125-023-06039-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/31/2023] [Indexed: 11/09/2023]
Abstract
AIMS/HYPOTHESIS Compromised pancreatic sympathetic innervation has been suggested as a factor involved in both immune-mediated beta cell destruction and endocrine dysregulation of pancreatic islets. To further explore these intriguing findings, new techniques for in vivo assessment of pancreatic innervation are required. This is a retrospective study that aimed to investigate whether the noradrenaline (norepinephrine) analogue 11C-hydroxy ephedrine (11C-HED) could be used for quantitative positron emission tomography (PET) imaging of the sympathetic innervation of the human pancreas. METHODS In 25 individuals with type 2 diabetes and 64 individuals without diabetes, all of whom had previously undergone 11C-HED-PET/CT because of pheochromocytoma or paraganglioma (or suspicion thereof), the 11C-HED standardised uptake value (SUVmean), 11C-HED specific binding index (SBI), pancreatic functional volume (FV, in ml), functional neuronal volume (FNV, calculated as SUVmean × FV), specific binding index with functional volume (SBI FV, calculated as SBI × FV) and attenuation on CT (HU) were investigated in the entire pancreas, and additionally in six separate anatomical pancreatic regions. RESULTS Generally, 11C-HED uptake in the pancreas was high, with marked individual variation, suggesting variability in sympathetic innervation. Moreover, pancreatic CT attenuation (HU) (p<0.001), 11C-HED SBI (p=0.0049) and SBI FV (p=0.0142) were lower in individuals with type 2 diabetes than in individuals without diabetes, whereas 11C-HED SUVmean (p=0.15), FV (p=0.73) and FNV (p=0.30) were similar. CONCLUSIONS/INTERPRETATION We demonstrate the feasibility of using 11C-HED-PET for non-invasive assessment of pancreatic sympathetic innervation in humans. These findings warrant further prospective evaluation, especially in individuals with theoretical defects in pancreatic sympathetic innervation, such as those with type 1 diabetes.
Collapse
Affiliation(s)
- Achyut Ram Vyakaranam
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden.
| | - Maryama M Mahamed
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Daniel Espes
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gustaf Christoffersson
- Science for Life Laboratory, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Anders Sundin
- Department of Surgical Sciences, Section of Radiology & Molecular Imaging, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Münzberg H, Berthoud HR, Neuhuber WL. Sensory spinal interoceptive pathways and energy balance regulation. Mol Metab 2023; 78:101817. [PMID: 37806487 PMCID: PMC10590858 DOI: 10.1016/j.molmet.2023.101817] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023] Open
Abstract
Interoception plays an important role in homeostatic regulation of energy intake and metabolism. Major interoceptive pathways include gut-to-brain and adipose tissue-to brain signaling via vagal sensory nerves and hormones, such as leptin. However, signaling via spinal sensory neurons is rapidly emerging as an additional important signaling pathway. Here we provide an in-depth review of the known anatomy and functions of spinal sensory pathways and discuss potential mechanisms relevant for energy balance homeostasis in health and disease. Because sensory innervation by dorsal root ganglia (DRG) neurons goes far beyond vagally innervated viscera and includes adipose tissue, skeletal muscle, and skin, it is in a position to provide much more complete metabolic information to the brain. Molecular and anatomical identification of function specific DRG neurons will be important steps in designing pharmacological and neuromodulation approaches to affect energy balance regulation in disease states such as obesity, diabetes, and cancer.
Collapse
Affiliation(s)
- Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | - Winfried L Neuhuber
- Institute for Anatomy and Cell Biology, Friedrich-Alexander University, Erlangen, Germany.
| |
Collapse
|
17
|
Bao J, Gao Z, Hu Y, Ye L, Wang L. Transient receptor potential vanilloid type 1: cardioprotective effects in diabetic models. Channels (Austin) 2023; 17:2281743. [PMID: 37983306 PMCID: PMC10761101 DOI: 10.1080/19336950.2023.2281743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
Cardiovascular disease, especially heart failure (HF) is the leading cause of death in patients with diabetes. Individuals with diabetes are prone to a special type of cardiomyopathy called diabetic cardiomyopathy (DCM), which cannot be explained by heart diseases such as hypertension or coronary artery disease, and can contribute to HF. Unfortunately, the current treatment strategy for diabetes-related cardiovascular complications is mainly to control blood glucose levels; nonetheless, the improvement of cardiac structure and function is not ideal. The transient receptor potential cation channel subfamily V member 1 (TRPV1), a nonselective cation channel, has been shown to be universally expressed in the cardiovascular system. Increasing evidence has shown that the activation of TRPV1 channel has a potential protective influence on the cardiovascular system. Numerous studies show that activating TRPV1 channels can improve the occurrence and progression of diabetes-related complications, including cardiomyopathy; however, the specific mechanisms and effects are unclear. In this review, we summarize that TRPV1 channel activation plays a protective role in the heart of diabetic models from oxidation/nitrification stress, mitochondrial function, endothelial function, inflammation, and cardiac energy metabolism to inhibit the occurrence and progression of DCM. Therefore, TRPV1 may become a latent target for the prevention and treatment of diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhicheng Gao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Miyamoto S, Kondo T, Maruyama K. Senso-immunology: the past, present, and future. J Biochem 2023; 174:305-315. [PMID: 37461198 DOI: 10.1093/jb/mvad052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 09/29/2023] Open
Abstract
Pain and mechanical stimulation are thought to be alarm systems that alert the brain to physical abnormalities. When we experience unpleasant feelings in infected or traumatized tissues, our awareness is directed to the afflicted region, prompting activities such as resting or licking the tissue. Despite extensive research into the molecular biology of nociceptors, it was unclear whether their role was limited to the generation and transmission of unpleasant feelings or whether they actively modulate the pathogenesis of infected or traumatized tissues. Recently, it has become clear how the sensory and immune systems interact with one another and share similar receptors and ligands to modify the pathogenesis of various diseases. In this paper, we summarize the mechanisms of crosstalk between the sensory and immune systems and the impact of this new interdisciplinary field, which should be dubbed 'senso-immunology,' on medical science.
Collapse
Affiliation(s)
- Satoshi Miyamoto
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 3N7, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Kondo
- Department of Biochemistry, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Hokkaido 060-8636, Japan
| | - Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Aichi 444-8787, Japan
| |
Collapse
|
19
|
Boahen A, Hu D, Adams MJ, Nicholls PK, Greene WK, Ma B. Bidirectional crosstalk between the peripheral nervous system and lymphoid tissues/organs. Front Immunol 2023; 14:1254054. [PMID: 37767094 PMCID: PMC10520967 DOI: 10.3389/fimmu.2023.1254054] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
The central nervous system (CNS) influences the immune system generally by regulating the systemic concentration of humoral substances (e.g., cortisol and epinephrine), whereas the peripheral nervous system (PNS) communicates specifically with the immune system according to local interactions/connections. An imbalance between the components of the PNS might contribute to pathogenesis and the further development of certain diseases. In this review, we have explored the "thread" (hardwiring) of the connections between the immune system (e.g., primary/secondary/tertiary lymphoid tissues/organs) and PNS (e.g., sensory, sympathetic, parasympathetic, and enteric nervous systems (ENS)) in health and disease in vitro and in vivo. Neuroimmune cell units provide an anatomical and physiological basis for bidirectional crosstalk between the PNS and the immune system in peripheral tissues, including lymphoid tissues and organs. These neuroimmune interactions/modulation studies might greatly contribute to a better understanding of the mechanisms through which the PNS possibly affects cellular and humoral-mediated immune responses or vice versa in health and diseases. Physical, chemical, pharmacological, and other manipulations of these neuroimmune interactions should bring about the development of practical therapeutic applications for certain neurological, neuroimmunological, infectious, inflammatory, and immunological disorders/diseases.
Collapse
Affiliation(s)
- Angela Boahen
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Seri-Kembangan, Selangor, Malaysia
| | - Dailun Hu
- Department of Pathogenic Biology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Murray J. Adams
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Philip K. Nicholls
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Wayne K. Greene
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| | - Bin Ma
- School of Medical, Molecular and Forensic Sciences, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
20
|
Sun M, Wan Y, Shi M, Meng ZX, Zeng W. Neural innervation in adipose tissue, gut, pancreas, and liver. LIFE METABOLISM 2023; 2:load022. [PMID: 39872245 PMCID: PMC11749697 DOI: 10.1093/lifemeta/load022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/22/2023] [Accepted: 06/05/2023] [Indexed: 01/30/2025]
Abstract
Efficient communication between the brain and peripheral organs is indispensable for regulating physiological function and maintaining energy homeostasis. The peripheral nervous system (PNS) in vertebrates, consisting of the autonomic and somatic nervous systems, bridges the peripheral organs and the central nervous system (CNS). Metabolic signals are processed by both vagal sensory nerves and somatosensory nerves. The CNS receives sensory inputs via ascending nerves, serves as the coordination and integration center, and subsequently controls internal organs and glands via descending nerves. The autonomic nervous system consists of sympathetic and parasympathetic branches that project peripheral nerves into various anatomical locations to regulate the energy balance. Sympathetic and parasympathetic nerves typically control the reflexive and involuntary functions in organs. In this review article, we outline the innervation of adipose tissue, gut, pancreas, and liver, to illustrate the neurobiological basis of central-peripheral interactions. We emphasize the importance of understanding the functional atlas of neural control of energy metabolism, and more importantly, provide potential avenues for further research in this area.
Collapse
Affiliation(s)
- Mengxue Sun
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yongwen Wan
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Mengjie Shi
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
21
|
Corral-Pujol M, Arpa B, Rosell-Mases E, Egia-Mendikute L, Mora C, Stratmann T, Sanchez A, Casanovas A, Esquerda JE, Mauricio D, Vives-Pi M, Verdaguer J. NOD mouse dorsal root ganglia display morphological and gene expression defects before and during autoimmune diabetes development. Front Endocrinol (Lausanne) 2023; 14:1176566. [PMID: 37334284 PMCID: PMC10272810 DOI: 10.3389/fendo.2023.1176566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction During the development of Autoimmune Diabetes (AD) an autoimmune attack against the Peripheral Nervous System occurs. To gain insight into this topic, analyses of Dorsal Root Ganglia (DRG) from Non-Obese Diabetic (NOD) mice were carried out. Methods Histopathological analysis by electron and optical microscopy in DRG samples, and mRNA expression analyzes by the microarray technique in DRG and blood leukocyte samples from NOD and C57BL/6 mice were performed. Results The results showed the formation of cytoplasmic vacuoles in DRG cells early in life that could be related to a neurodegenerative process. In view of these results, mRNA expression analyses were conducted to determine the cause and/or the molecules involved in this suspected disorder. The results showed that DRG cells from NOD mice have alterations in the transcription of a wide range of genes, which explain the previously observed alterations. In addition, differences in the transcription genes in white blood cells were also detected. Discussion Taken together, these results indicate that functional defects are not only seen in beta cells but also in DRG in NOD mice. These results also indicate that these defects are not a consequence of the autoimmune process that takes place in NOD mice and suggest that they may be involved as triggers for its development.
Collapse
Affiliation(s)
- Marta Corral-Pujol
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Berta Arpa
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Estela Rosell-Mases
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Leire Egia-Mendikute
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Conchi Mora
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
| | - Thomas Stratmann
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Alex Sanchez
- Genetics, Microbiology and Statistics Department, Universitat de Barcelona, Barcelona, Spain
- Statistics and Bioinformatics Unit, Vall d’Hebron Institut de Recerca, Barcelona, Spain
| | - Anna Casanovas
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Josep Enric Esquerda
- Patologia Neuromuscular Experimental Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Didac Mauricio
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), Barcelona, Spain
- Faculty of Medicine, Central University of Catalonia, Vic, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Vives-Pi
- Immunology Department, Germans Trias i Pujol Research Institute, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Faculty of Medicine, Autonomous University of Barcelona, Cerdanyola del Vallès, Spain
| | - Joan Verdaguer
- Immunology Unit, Department of Experimental Medicine, Faculty of Medicine, University of Lleida and IRBLleida, Lleida, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Wang Y, Ye L. Somatosensory innervation of adipose tissues. Physiol Behav 2023; 265:114174. [PMID: 36965573 PMCID: PMC11537203 DOI: 10.1016/j.physbeh.2023.114174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
The increasing prevalence of obesity and type 2 diabetes has led to a greater interest in adipose tissue physiology. Adipose tissue is now understood as an organ with endocrine and thermogenic capacities in addition to its role in fat storage. It plays a critical role in systemic metabolism and energy regulation, and its activity is tightly regulated by the nervous system. Fat is now recognized to receive sympathetic innervation, which transmits information from the brain, as well as sensory innervation, which sends information into the brain. The role of sympathetic innervation in adipose tissue has been extensively studied. However, the extent and the functional significance of sensory innervation have long been unclear. Recent studies have started to reveal that sensory neurons robustly innervate adipose tissue and play an important role in regulating fat activity. This brief review will discuss both historical evidence and recent advances, as well as important remaining questions about the sensory innervation of adipose tissue.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Li Ye
- Department of Neuroscience and Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
23
|
TRPV1 is involved in abdominal hyperalgesia in a mouse model of lipopolysaccharide-induced peritonitis and influences the immune response via peripheral noradrenergic neurons. Life Sci 2023; 317:121472. [PMID: 36750138 DOI: 10.1016/j.lfs.2023.121472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 01/12/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
AIMS The transient receptor potential vanilloid subfamily 1 (TRPV1) not only plays a role as a nociceptor but also has some regulatory effects on the immune system. We investigated the effects of TRPV1 on abdominal pain and the immune system in lipopolysaccharide (LPS)-induced peritonitis and the association between TRPV1 and peripheral noradrenergic neurons. MAIN METHODS Experiments were performed in 8- to 14-week-old male wild-type (WT) and TRPV1 knockout (KO) mice. The mice were intraperitoneally injected with a non-lethal dose of LPS. Pain assessment and investigation of changes in the immune system were performed. Denervation of sympathetic nerves and the noradrenergic splenic nerve was induced by intraperitoneal administration of 6-hydroxydopamine. KEY FINDINGS The levels of serum cytokines were not significantly different in WT mice and TRPV1 KO mice. Abdominal mechanical hyperalgesia was greater in WT mice than in TRPV1 KO mice from 6 h to 3 days. The numbers of macrophages, neutrophils, dendritic cells, and CD4 T cells in the spleens of TRPV1 KO mice were significantly increased compared to those in WT mice 4 days after LPS administration. By noradrenergic denervation, the numbers of those cells in WT mice increased to levels comparable to those in TRPV1 KO mice. SIGNIFICANCE In LPS-induced peritonitis, abdominal inflammatory pain was transmitted via TRPV1. In addition, TRPV1 had an anti-inflammatory effect on the spleen in the late phase of peritonitis. This anti-inflammatory effect was thought to be mediated by activation of the sympathetic nervous system and/or noradrenergic splenic nerve induced by TRPV1 activation.
Collapse
|
24
|
Mandal SK, Rath SK, Logesh R, Mishra SK, Devkota HP, Das N. Capsicum annuum L. and its bioactive constituents: A critical review of a traditional culinary spice in terms of its modern pharmacological potentials with toxicological issues. Phytother Res 2023; 37:965-1002. [PMID: 36255140 DOI: 10.1002/ptr.7660] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/07/2022] [Accepted: 09/27/2022] [Indexed: 11/12/2022]
Abstract
Capsicum annuum L., commonly known as chili pepper, is used as an important spice globally and as a crude drug in many traditional medicine systems. The fruits of C. annuum have been used as a tonic, antiseptic, and stimulating agent, to treat dyspepsia, appetites, and flatulence, and to improve digestion and circulation. The article aims to critically review the phytochemical and pharmacological properties of C. annuum and its major compounds. Capsaicin, dihydrocapsaicin, and some carotenoids are reported as the major active compounds with several pharmacological potentials especially as anticancer and cardioprotectant. The anticancer effect of capsaicinoids is mainly mediated through mechanisms involving the interaction of Ca2+ -dependent activation of the MAPK pathway, suppression of NOX-dependent reactive oxygen species generation, and p53-mediated activation of mitochondrial apoptosis in cancer cells. Similarly, the cardioprotective effects of capsaicinoids are mediated through their interaction with cellular transient receptor potential vanilloid 1 channel, and restoration of calcitonin gene-related peptide via Ca2+ -dependent release of neuropeptides and suppression of bradykinin. In conclusion, this comprehensive review presents detailed information about the traditional uses, phytochemistry, and pharmacology of major bioactive principles of C. annuum with special emphasis on anticancer, cardioprotective effects, and plausible toxic adversities along with food safety.
Collapse
Affiliation(s)
- Sudip Kumar Mandal
- Department of Pharmaceutical Chemistry, Dr. B. C. Roy College of Pharmacy and AHS, Durgapur, India
| | - Santosh Kumar Rath
- School of Pharmaceuticals and Population Health Informatics, Faculty of Pharmacy, DIT University, Dehradun, India
| | - Rajan Logesh
- TIFAC CORE in Herbal Drugs, Department of Pharmacognosy, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Rockland's, Ooty, India
| | | | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Niranjan Das
- Department of Chemistry, Ramthakur College, Agartala, India
| |
Collapse
|
25
|
Kokabi F, Ebrahimi S, Mirzavi F, Ghiasi Nooghabi N, Hashemi SF, Hashemy SI. The neuropeptide substance P/neurokinin-1 receptor system and diabetes: From mechanism to therapy. Biofactors 2023. [PMID: 36651605 DOI: 10.1002/biof.1935] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
Diabetes is a significant public health issue known as the world's fastest-growing disease condition. It is characterized by persistent hyperglycemia and subsequent chronic complications leading to organ dysfunction and, ultimately, the failure of target organs. Substance P (SP) is an undecapeptide that belongs to the family of tachykinin (TK) peptides. The SP-mediated activation of the neurokinin 1 receptor (NK1R) regulates many pathophysiological processes in the body. There is also a relation between the SP/NK1R system and diabetic processes. Importantly, deregulated expression of SP has been reported in diabetes and diabetes-associated chronic complications. SP can induce both diabetogenic and antidiabetogenic effects and thus affect the pathology of diabetes destructively or protectively. Here, we review the current knowledge of the functional relevance of the SP/NK1R system in diabetes pathogenesis and its exploitation for diabetes therapy. A comprehensive understanding of the role of the SP/NK1R system in diabetes is expected to shed further light on developing new therapeutic possibilities for diabetes and its associated chronic conditions.
Collapse
Affiliation(s)
- Fariba Kokabi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safieh Ebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | | | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
26
|
Szallasi A. Dietary Capsaicin: A Spicy Way to Improve Cardio-Metabolic Health? Biomolecules 2022; 12:biom12121783. [PMID: 36551210 PMCID: PMC9775666 DOI: 10.3390/biom12121783] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Today's sedentary lifestyle with too much food and too little exercise has made metabolic syndrome a pandemic. Metabolic syndrome is a major risk factor for type-2 diabetes and cardiovascular disease. New knowledge of medical and nutraceutical intervention in the early stages of metabolic syndrome is central to prevent these deadly complications. People who eat chili pepper on a regular basis seem to stay healthier and live longer than those who do not. Animal experiments suggest a therapeutic potential for dietary capsaicin, the active principle in hot chili pepper, to reduce the risk of developing metabolic syndrome. This is an attractive theory since capsaicin has been a culinary staple for thousands of years, and is generally deemed safe when consumed in hedonically acceptable doses. The broad expression of the capsaicin receptor TRPV1 in metabolically active tissues lends experimental support to this theory. This review critically evaluates the available experimental and clinical evidence for and against dietary capsaicin being an effective dietary means to improve cardio-metabolic health. It comes to the conclusion that although a chili pepper-rich diet is associated with a reduced risk of dying due to cardiovascular disease, dietary capsaicin has no clear effect on blood glucose or lipid profiles. Therefore, the reduced mortality risk may reflect the beneficial action of digested capsaicin on gut microbiota.
Collapse
Affiliation(s)
- Arpad Szallasi
- Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| |
Collapse
|
27
|
Bougnères P, Le Fur S, Valleron AJ. Early varicella infection is associated with a delayed onset of childhood type 1 diabetes. DIABETES & METABOLISM 2022; 48:101394. [PMID: 36170944 DOI: 10.1016/j.diabet.2022.101394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Affiliation(s)
- Pierre Bougnères
- Institut National de la Santé et de la Recherche Médicale (U1195), Le Kremlin-Bicêtre, France; Paris-Saclay University, France.
| | - Sophie Le Fur
- Institut National de la Santé et de la Recherche Médicale (U1195), Le Kremlin-Bicêtre, France
| | - Alain-Jacques Valleron
- Institut National de la Santé et de la Recherche Médicale (U1195), Le Kremlin-Bicêtre, France
| |
Collapse
|
28
|
Liu C, Miao R, Raza F, Qian H, Tian X. Research progress and challenges of TRPV1 channel modulators as a prospective therapy for diabetic neuropathic pain. Eur J Med Chem 2022; 245:114893. [DOI: 10.1016/j.ejmech.2022.114893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
|
29
|
Peng G, Tang X, Gui Y, Yang J, Ye L, Wu L, Ding YH, Wang L. Transient receptor potential vanilloid subtype 1: A potential therapeutic target for fibrotic diseases. Front Physiol 2022; 13:951980. [PMID: 36045746 PMCID: PMC9420870 DOI: 10.3389/fphys.2022.951980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
The transient receptor potential vanilloid subtype 1 (TRPV1), belonging to the TRPV channel family, is a non-selective, calcium-dependent, cation channel implicated in several pathophysiological processes. Collagen, an extracellular matrix component, can accumulate under pathological conditions and may lead to the destruction of tissue structure, organ dysfunction, and organ failure. Increasing evidence indicates that TRPV1 plays a role in the development and occurrence of fibrotic diseases, including myocardial, renal, pancreatic, and corneal fibrosis. However, the mechanism by which TRPV1 regulates fibrosis remains unclear. This review highlights the comprehensive role played by TRPV1 in regulating pro-fibrotic processes, the potential of TRPV1 as a therapeutic target in fibrotic diseases, as well as the different signaling pathways associated with TRPV1 and fibrosis.
Collapse
Affiliation(s)
- Guangxin Peng
- Zhejiang University of Technology, Hangzhou, China
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaoling Tang
- Zhejiang University of Technology, Hangzhou, China
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yang Gui
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jing Yang
- Zhejiang University of Technology, Hangzhou, China
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lifang Ye
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liuyang Wu
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ya hui Ding
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Lihong Wang
- Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Lihong Wang,
| |
Collapse
|
30
|
Wang S, Nie X, Siddiqui Y, Wang X, Arora V, Fan X, Thumbigere-Math V, Chung M. Nociceptor Neurons Magnify Host Responses to Aggravate Periodontitis. J Dent Res 2022; 101:812-820. [PMID: 35086367 PMCID: PMC9210118 DOI: 10.1177/00220345211069956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Periodontitis is a highly prevalent chronic inflammatory disease that progressively destroys the structures supporting teeth, leading to tooth loss. Periodontal tissue is innervated by abundant pain-sensing primary afferents expressing neuropeptides and transient receptor potential vanilloid 1 (TRPV1). However, the roles of nociceptive nerves in periodontitis and bone destruction are controversial. The placement of ligature around the maxillary second molar or the oral inoculation of pathogenic bacteria induced alveolar bone destruction in mice. Chemical ablation of nociceptive neurons in the trigeminal ganglia achieved by intraganglionic injection of resiniferatoxin decreased bone loss in mouse models of experimental periodontitis. Consistently, ablation of nociceptive neurons decreased the number of osteoclasts in alveolar bone under periodontitis. The roles of nociceptors were also determined by the functional inhibition of TRPV1-expressing trigeminal afferents using an inhibitory designer receptor exclusively activated by designer drugs (DREADD) receptor. Noninvasive chemogenetic functional silencing of TRPV1-expressing trigeminal afferents not only decreased induction but also reduced the progression of bone loss in periodontitis. The infiltration of leukocytes and neutrophils to the periodontium increased at the site of ligature, which was accompanied by increased amount of proinflammatory cytokines, such as receptor activator of nuclear factor κΒ ligand, tumor necrosis factor, and interleukin 1β. The extents of increase in immune cell infiltration and cytokines were significantly lower in mice with nociceptor ablation. In contrast, the ablation of nociceptors did not alter the periodontal microbiome under the conditions of control and periodontitis. Altogether, these results indicate that TRPV1-expressing afferents increase bone destruction in periodontitis by promoting hyperactive host responses in the periodontium. We suggest that specific targeting of neuroimmune and neuroskeletal regulation can offer promising therapeutic targets for periodontitis supplementing conventional treatments.
Collapse
Affiliation(s)
- S. Wang
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - X. Nie
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Y. Siddiqui
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - X. Wang
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - V. Arora
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - X. Fan
- Department of Microbiology and Immunology, Flow Cytometry Shared Service, University of Maryland School of Medicine, Baltimore, MD, USA
| | - V. Thumbigere-Math
- Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - M.K. Chung
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| |
Collapse
|
31
|
Hampton RF, Jimenez-Gonzalez M, Stanley SA. Unravelling innervation of pancreatic islets. Diabetologia 2022; 65:1069-1084. [PMID: 35348820 PMCID: PMC9205575 DOI: 10.1007/s00125-022-05691-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/08/2022] [Indexed: 01/05/2023]
Abstract
The central and peripheral nervous systems play critical roles in regulating pancreatic islet function and glucose metabolism. Over the last century, in vitro and in vivo studies along with examination of human pancreas samples have revealed the structure of islet innervation, investigated the contribution of sympathetic, parasympathetic and sensory neural pathways to glucose control, and begun to determine how the structure and function of pancreatic nerves are disrupted in metabolic disease. Now, state-of-the art techniques such as 3D imaging of pancreatic innervation and targeted in vivo neuromodulation provide further insights into the anatomy and physiological roles of islet innervation. Here, we provide a summary of the published work on the anatomy of pancreatic islet innervation, its roles, and evidence for disordered islet innervation in metabolic disease. Finally, we discuss the possibilities offered by new technologies to increase our knowledge of islet innervation and its contributions to metabolic regulation.
Collapse
Affiliation(s)
- Rollie F Hampton
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Jimenez-Gonzalez
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
32
|
Liu Y, Lyu Y, Wang H. TRP Channels as Molecular Targets to Relieve Endocrine-Related Diseases. Front Mol Biosci 2022; 9:895814. [PMID: 35573736 PMCID: PMC9095829 DOI: 10.3389/fmolb.2022.895814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 12/03/2022] Open
Abstract
Transient receptor potential (TRP) channels are polymodal channels capable of sensing environmental stimuli, which are widely expressed on the plasma membrane of cells and play an essential role in the physiological or pathological processes of cells as sensors. TRPs often form functional homo- or heterotetramers that act as cation channels to flow Na+ and Ca2+, change membrane potential and [Ca2+]i (cytosolic [Ca2+]), and change protein expression levels, channel attributes, and regulatory factors. Under normal circumstances, various TRP channels respond to intracellular and extracellular stimuli such as temperature, pH, osmotic pressure, chemicals, cytokines, and cell damage and depletion of Ca2+ reserves. As cation transport channels and physical and chemical stimulation receptors, TRPs play an important role in regulating secretion, interfering with cell proliferation, and affecting neural activity in these glands and their adenocarcinoma cells. Many studies have proved that TRPs are widely distributed in the pancreas, adrenal gland, and other glands. This article reviews the specific regulatory mechanisms of various TRP channels in some common glands (pancreas, salivary gland, lacrimal gland, adrenal gland, mammary gland, gallbladder, and sweat gland).
Collapse
|
33
|
Araújo MC, Soczek SHS, Pontes JP, Marques LAC, Santos GS, Simão G, Bueno LR, Maria-Ferreira D, Muscará MN, Fernandes ES. An Overview of the TRP-Oxidative Stress Axis in Metabolic Syndrome: Insights for Novel Therapeutic Approaches. Cells 2022; 11:cells11081292. [PMID: 35455971 PMCID: PMC9030853 DOI: 10.3390/cells11081292] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MS) is a complex pathology characterized by visceral adiposity, insulin resistance, arterial hypertension, and dyslipidaemia. It has become a global epidemic associated with increased consumption of high-calorie, low-fibre food and sedentary habits. Some of its underlying mechanisms have been identified, with hypoadiponectinemia, inflammation and oxidative stress as important factors for MS establishment and progression. Alterations in adipokine levels may favour glucotoxicity and lipotoxicity which, in turn, contribute to inflammation and cellular stress responses within the adipose, pancreatic and liver tissues, in addition to hepatic steatosis. The multiple mechanisms of MS make its clinical management difficult, involving both non-pharmacological and pharmacological interventions. Transient receptor potential (TRP) channels are non-selective calcium channels involved in a plethora of physiological events, including energy balance, inflammation and oxidative stress. Evidence from animal models of disease has contributed to identify their specific contributions to MS and may help to tailor clinical trials for the disease. In this context, the oxidative stress sensors TRPV1, TRPA1 and TRPC5, play major roles in regulating inflammatory responses, thermogenesis and energy expenditure. Here, the interplay between these TRP channels and oxidative stress in MS is discussed in the light of novel therapies to treat this syndrome.
Collapse
Affiliation(s)
- Mizael C. Araújo
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Suzany H. S. Soczek
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Jaqueline P. Pontes
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 565085-080, MA, Brazil;
| | - Leonardo A. C. Marques
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Gabriela S. Santos
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Laryssa R. Bueno
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Marcelo N. Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Elizabeth S. Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
- Correspondence:
| |
Collapse
|
34
|
Nociceptor-derived Reg3γ prevents endotoxic death by targeting kynurenine pathway in microglia. Cell Rep 2022; 38:110462. [PMID: 35263589 DOI: 10.1016/j.celrep.2022.110462] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 11/21/2022] Open
Abstract
Nociceptors can fine-tune local or systemic immunity, but the mechanisms of nociceptive modulation in endotoxic death remain largely unknown. Here, we identified C-type lectin Reg3γ as a nociceptor-enriched hormone that protects the host from endotoxic death. During endotoxemia, nociceptor-derived Reg3γ penetrates the brain and suppresses the expression of microglial indoleamine dioxygenase 1, a critical enzyme of the kynurenine pathway, via the Extl3-Bcl10 axis. Endotoxin-administered nociceptor-null mice and nociceptor-specific Reg3γ-deficient mice exhibit a high mortality rate accompanied by decreased brain HK1 phosphorylation and ATP production despite normal peripheral inflammation. Such metabolic arrest is only observed in the brain, and aberrant production of brain quinolinic acid, a neurotoxic metabolite of the kynurenine pathway, causes HK1 suppression. Strikingly, the central administration of Reg3γ protects mice from endotoxic death by enhancing brain ATP production. By identifying nociceptor-derived Reg3γ as a microglia-targeted hormone, this study provides insights into the understanding of tolerance to endotoxic death.
Collapse
|
35
|
Mirzadeh Z, Faber CL, Schwartz MW. Central Nervous System Control of Glucose Homeostasis: A Therapeutic Target for Type 2 Diabetes? Annu Rev Pharmacol Toxicol 2022; 62:55-84. [PMID: 34990204 PMCID: PMC8900291 DOI: 10.1146/annurev-pharmtox-052220-010446] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Historically, pancreatic islet beta cells have been viewed as principal regulators of glycemia, with type 2 diabetes (T2D) resulting when insulin secretion fails to compensate for peripheral tissue insulin resistance. However, glycemia is also regulated by insulin-independent mechanisms that are dysregulated in T2D. Based on evidence supporting its role both in adaptive coupling of insulin secretion to changes in insulin sensitivity and in the regulation of insulin-independent glucose disposal, the central nervous system (CNS) has emerged as a fundamental player in glucose homeostasis. Here, we review and expand upon an integrative model wherein the CNS, together with the islet, establishes and maintains the defended level of glycemia. We discuss the implications of this model for understanding both normal glucose homeostasis and T2D pathogenesis and highlight centrally targeted therapeutic approaches with the potential to restore normoglycemia to patients with T2D.
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Ivy Brain Tumor Center, Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA;
| | - Chelsea L Faber
- Ivy Brain Tumor Center, Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona 85013, USA;
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington 98109, USA;
| | - Michael W Schwartz
- UW Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, Washington 98109, USA;
| |
Collapse
|
36
|
Lian J, Casari I, Falasca M. Modulatory role of the endocannabinoidome in the pathophysiology of the gastrointestinal tract. Pharmacol Res 2021; 175:106025. [PMID: 34883211 DOI: 10.1016/j.phrs.2021.106025] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/29/2021] [Accepted: 12/05/2021] [Indexed: 12/13/2022]
Abstract
Originating from Eastern Asia, the plant Cannabis sativa has been used for centuries as a medicinal treatment. The unwanted psychotropic effects of one of its major components, Δ9-tetrahydrocannabinol, discouraged its therapeutic employment until, recently, the discovery of cannabinoids receptors and their endogenous ligands endocannabinoids reignited the interest. The endocannabinoid system has lately been found to play an important role in the maintenance of human health, both centrally and peripherally. However, the initial idea of the endocannabinoid system structure has been quickly understood to be too simplistic and, as new receptors, mediators, and enzymes have been discovered to participate in a complex relationship, the new, more comprehensive term "expanded endocannabinoid system" or "endocannabinoidome", has taken over. The discovery of other endocannabinoid-like receptors, such as the G protein-coupled receptor 119 and G protein-coupled receptor 55, has opened the way to the development of potential therapeutic targets for the treatment of various metabolic disorders. In addition, recent findings have also provided evidence suggesting the potential therapeutic link between the endocannabinoidome and various inflammatory-based gut diseases, such as inflammatory bowel disease and cancer. This review will provide an introduction to the endocannabinoidome, focusing on its modulatory role in the gastrointestinal tract and on the interest generated by the link between gut microbiota, the endocannabinoid system and metabolic diseases such as inflammatory bowel disease, type-2 diabetes and obesity. In addition, we will look at the potential novel aspects and benefits of drugs targeting the endocannabinoid system.
Collapse
Affiliation(s)
- Jerome Lian
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Ilaria Casari
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia
| | - Marco Falasca
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia 6102, Australia.
| |
Collapse
|
37
|
Smoak P, Burke SJ, Collier JJ. Botanical Interventions to Improve Glucose Control and Options for Diabetes Therapy. SN COMPREHENSIVE CLINICAL MEDICINE 2021; 3:2465-2491. [PMID: 35098034 PMCID: PMC8796700 DOI: 10.1007/s42399-021-01034-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Diabetes mellitus is a major public health problem worldwide. This endocrine disease is clustered into distinct subtypes based on the route of development, with the most common forms associated with either autoimmunity (T1DM) or obesity (T2DM). A shared hallmark of both major forms of diabetes is a reduction in function (insulin secretion) or mass (cell number) of the pancreatic islet beta-cell. Diminutions in both mass and function are often present. A wide assortment of plants have been used historically to reduce the pathological features associated with diabetes. In this review, we provide an organized viewpoint focused around the phytochemicals and herbal extracts investigated using various preclinical and clinical study designs. In some cases, crude extracts were examined directly, and in others, purified compounds were explored for their possible therapeutic efficacy. A subset of these studies compared the botanical product with standard of care prescribed drugs. Finally, we note that botanical formulations are likely suspects for future drug discovery and refinement into class(es) of compounds that have either direct or adjuvant therapeutic benefit.
Collapse
Affiliation(s)
- Peter Smoak
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | - Susan J. Burke
- Immunogenetics Laboratory, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, LA 70808 Baton Rouge, USA
| | - J. Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| |
Collapse
|
38
|
Costa PAC, Silva WN, Prazeres PHDM, Picoli CC, Guardia GDA, Costa AC, Oliveira MA, Guimarães PPG, Gonçalves R, Pinto MCX, Amorim JH, Azevedo VAC, Resende RR, Russo RC, Cunha TM, Galante PAF, Mintz A, Birbrair A. Chemogenetic modulation of sensory neurons reveals their regulating role in melanoma progression. Acta Neuropathol Commun 2021; 9:183. [PMID: 34784974 PMCID: PMC8594104 DOI: 10.1186/s40478-021-01273-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/10/2021] [Indexed: 02/08/2023] Open
Abstract
Sensory neurons have recently emerged as components of the tumor microenvironment. Nevertheless, whether sensory neuronal activity is important for tumor progression remains unknown. Here we used Designer Receptors Exclusively Activated by a Designer Drug (DREADD) technology to inhibit or activate sensory neurons' firing within the melanoma tumor. Melanoma growth and angiogenesis were accelerated following inhibition of sensory neurons' activity and were reduced following overstimulation of these neurons. Sensory neuron-specific overactivation also induced a boost in the immune surveillance by increasing tumor-infiltrating anti-tumor lymphocytes, while reducing immune-suppressor cells. In humans, a retrospective in silico analysis of melanoma biopsies revealed that increased expression of sensory neurons-related genes within melanoma was associated with improved survival. These findings suggest that sensory innervations regulate melanoma progression, indicating that manipulation of sensory neurons' activity may provide a valuable tool to improve melanoma patients' outcomes.
Collapse
Affiliation(s)
- Pedro A C Costa
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Walison N Silva
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Pedro H D M Prazeres
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Caroline C Picoli
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | | | - Alinne C Costa
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Mariana A Oliveira
- Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Pedro P G Guimarães
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Ricardo Gonçalves
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Mauro C X Pinto
- Departamento de Farmacologia, Universidade Federal de Goiás, Goiânia, GO, Brasil
| | - Jaime H Amorim
- Centro das Ciências Biológicas e da Saúde, Universidade Federal do Oeste da Bahia, Barreiras, BA, Brasil
| | - Vasco A C Azevedo
- Departamento de Genetica, Ecologia e Evolucao, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Rodrigo R Resende
- Departamento de Bioquimica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Remo C Russo
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - Thiago M Cunha
- Departamento de Farmacologia, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Pedro A F Galante
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, Sao Paulo, SP, Brasil
| | - Akiva Mintz
- Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Alexander Birbrair
- Departamento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.
- Department of Radiology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
39
|
Xu T, Yu Z, Liu Y, Lu M, Gong M, Li Q, Xia Y, Xu B. Hypoglycemic Effect of Electroacupuncture at ST25 Through Neural Regulation of the Pancreatic Intrinsic Nervous System. Mol Neurobiol 2021; 59:703-716. [PMID: 34757591 PMCID: PMC8786791 DOI: 10.1007/s12035-021-02609-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023]
Abstract
Electroacupuncture (EA) is considered to have potential antidiabetic effects; however, the role of the pancreatic intrinsic nervous system (PINS) in EA-induced amelioration of type 2 diabetes (T2DM) remains unclear. Therefore, we investigated whether EA at ST25 exerts any beneficial effects on insulin resistance (IR), inflammation severity, and pancreatic β cell function via the PINS in a rat model of a high-fat diet-streptozotocin (HFD/STZ)-induced diabetes. To this end, Sprague Dawley rats were fed with HFD to induce IR, followed by STZ (35 mg/kg, i.p.) injection to establish the T2DM model. After hyperglycemia was confirmed as fasting glucose level > 16.7 mmol/L, the rats were treated with EA (2 mA, 2/15 Hz) for the next 28 days. Model rats showed increased serum glucose, insulin, IR, and TNF-α levels with a concomitant decrease in β cell function. Microscopy examination of the pancreas revealed pathological changes in islets, which reverted to near-normal levels after EA at ST25. EA improved islet cell morphology by increasing islet area and reducing vacuolation. EA at ST25 decreased transient receptor potential vanilloid 1 (TRPV1) and increased substance P (SP) and calcitonin gene-related peptide (CGRP) expression. Subsequently, insulin secretion decreased and impaired pancreatic endocrine function was restored through the TRPV1 channel (SP/CGRP)-insulin circuit. EA increased choline acetyltransferase and neuropeptide Y expression and controlled inflammation. It also enhanced the cocaine and amphetamine-regulated transcript prepropeptide expression and promoted glucagon-like peptide-1 secretion. Additionally, the electrophysiological activity of PINS during acupuncture (2.71 ± 1.72 Hz) was significantly increased compared to the pre-acupuncture frequency (0.32 ± 0.37 Hz, P < 0.05). Thus, our study demonstrated the beneficial effect of EA on β cell dysfunction via the PINS in rat models of HFD-STZ-induced T2DM.
Collapse
Affiliation(s)
- Tiancheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Mengjiang Lu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Meirong Gong
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qian Li
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Youbing Xia
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China. .,Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
40
|
Zhang S, Tang L, Xu F, Hui Y, Lu H, Liu X. TRPV1 Receptor-Mediated Hypoglycemic Mechanism of Capsaicin in Streptozotocin-Induced Diabetic Rats. Front Nutr 2021; 8:750355. [PMID: 34692753 PMCID: PMC8526734 DOI: 10.3389/fnut.2021.750355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Our previous research showed that capsaicin exhibits hypoglycemic effects by activating the transient receptor potential vanilloid 1 (TRPV1) channel in diabetic rats. Interestingly, capsiate was also able to activate the TRPV1 channel, but with a non-significant hypoglycemic effect. This study aimed to investigate the effect of capsaicin on the glycometabolism of streptozotocin (STZ)-induced diabetic rats by blocking the TRPV1 channel. After a 4-week capsaicin treatment (6 mg/kg·bw), the serum insulin level of STZ-induced diabetic rats increased from 15.2 to 22.1 mIU/L, the content of hepatic glycogen and muscle glycogen increased by 81.2 and 20.2%, respectively, and the blood glucose level decreased significantly from 19.3 to 14.7 mmol/L. When the TRPV1 channel was blocked, capsaicin lost the above-mentioned effects, and the hypoglycemic effect was no longer significant. It was concluded that a combined up-regulation of both TRPV1 receptors and pancreatic duodenal homeobox-1 (PDX-1) led to the hypoglycemic effect of capsaicin, which partially explains our previous observation: capsiate activating TRPV1 without showing a significant hypoglycemic effect was due to the lack of a significant up-regulation of PDX-1. Based on the experimental results, we speculated that two signaling pathways [TRPV1-(PDX1)-(GLUT2/GK) and TRPV1-(PDX-1)-(IRS1/2)] exist in the pancreas of STZ-induced diabetic rats.
Collapse
Affiliation(s)
- Shiqi Zhang
- College of Food Science and Engineering, Lingnan Normal University, Zhanjiang, China
| | - Lanlan Tang
- College of Food Science, Southwest University, Chongqing, China
| | - Fanshu Xu
- Department of Cell and System Biology, Faculty of Arts and Science, University of Toronto, Toronto, ON, Canada
| | - Yonghai Hui
- College of Food Science and Engineering, Lingnan Normal University, Zhanjiang, China
| | - Hongjia Lu
- College of Landscape Architecture and Life Science/Institute of Special Plants, Chongqing University of Arts and Science, Chongqing, China
| | - Xiong Liu
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
41
|
Neuro-immune-metabolism: The tripod system of homeostasis. Immunol Lett 2021; 240:77-97. [PMID: 34655659 DOI: 10.1016/j.imlet.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 11/20/2022]
Abstract
Homeostatic regulation of cellular and molecular processes is essential for the efficient physiological functioning of body organs. It requires an intricate balance of several networks throughout the body, most notable being the nervous, immune and metabolic systems. Several studies have reported the interactions between neuro-immune, immune-metabolic and neuro-metabolic pathways. Current review aims to integrate the information and show that neuro, immune and metabolic systems form the triumvirate of homeostasis. It focuses on the cellular and molecular interactions occurring in the extremities and intestine, which are innervated by the peripheral nervous system and for the intestine in particular the enteric nervous system. While the interdependence of neuro-immune-metabolic pathways provides a fallback mechanism in case of disruption of homeostasis, in chronic pathologies of continued disequilibrium, the collapse of one system spreads to the other interacting networks as well. Current review illustrates this domino-effect using diabetes as the main example. Together, this review attempts to provide a holistic picture of the integrated network of neuro-immune-metabolism and attempts to broaden the outlook when devising a scientific study or a treatment strategy.
Collapse
|
42
|
Lkhagvasuren B, Mee-Inta O, Zhao ZW, Hiramoto T, Boldbaatar D, Kuo YM. Pancreas-Brain Crosstalk. Front Neuroanat 2021; 15:691777. [PMID: 34354571 PMCID: PMC8329585 DOI: 10.3389/fnana.2021.691777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
The neural regulation of glucose homeostasis in normal and challenged conditions involves the modulation of pancreatic islet-cell function. Compromising the pancreas innervation causes islet autoimmunity in type 1 diabetes and islet cell dysfunction in type 2 diabetes. However, despite the richly innervated nature of the pancreas, islet innervation remains ill-defined. Here, we review the neuroanatomical and humoral basis of the cross-talk between the endocrine pancreas and autonomic and sensory neurons. Identifying the neurocircuitry and neurochemistry of the neuro-insular network would provide clues to neuromodulation-based approaches for the prevention and treatment of diabetes and obesity.
Collapse
Affiliation(s)
- Battuvshin Lkhagvasuren
- Brain Science Institute, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Onanong Mee-Inta
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Zi-Wei Zhao
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Tetsuya Hiramoto
- Department of Psychosomatic Medicine, Fukuoka Hospital, National Hospital Organization, Fukuoka, Japan
| | - Damdindorj Boldbaatar
- Brain Science Institute, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine, Tainan, Taiwan.,Department of Cell Biology and Anatomy, National Cheng Kung University College of Medicine, Tainan, Taiwan
| |
Collapse
|
43
|
Abstract
Transient receptor potential vanilloid subfamily member 1 (TRPV1) is a nonselective cation channel, that is mainly distributed in sensory nerve endings and can release a variety of neurotransmitters after activation. Early studies showed that it mainly conducts pain sensation, but research has demonstrated that it also plays an important role in cardiovascular diseases. Notably, in atherosclerosis, the activation of TRPV1 can regulate lipid metabolism, reduce foam cell formation, protect endothelial cells, inhibit smooth muscle cell proliferation and inhibit inflammation and oxidation. In this review, the role of the TRPV1 channel in atherosclerosis was discussed to provide new ideas for the prevention and treatment of atherosclerotic diseases.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Medicine, Qingdao University, Qingdao, China.,Zhejiang Provincial People's Hospital, Qingdao University, Hangzhou, China
| | - Lifang Ye
- Zhejiang Provincial People's Hospital, Qingdao University, Hangzhou, China
| | - Qinggang Zhang
- Zhejiang Provincial People's Hospital, Qingdao University, Hangzhou, China
| | - Fei Wu
- Zhejiang Provincial People's Hospital, Qingdao University, Hangzhou, China
| | - Lihong Wang
- Zhejiang Provincial People's Hospital, Qingdao University, Hangzhou, China
| |
Collapse
|
44
|
Gladkikh IN, Sintsova OV, Leychenko EV, Kozlov SA. TRPV1 Ion Channel: Structural Features, Activity Modulators, and Therapeutic Potential. BIOCHEMISTRY (MOSCOW) 2021; 86:S50-S70. [PMID: 33827400 DOI: 10.1134/s0006297921140054] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Although TRPV1 ion channel has been attracting researchers' attention for many years, its functions in animal organisms, the principles of regulation, and the involvement in pathological processes have not yet been fully clarified. Mutagenesis experiments and structural studies have identified the structural features of the channel and binding sites for its numerous ligands; however, these studies are far from conclusion. This review summarizes recent achievements in the TRPV1 research with special focus on structural and functional studies of the channel and on its ligands, which are extremely diverse in their nature and interaction specificity to TRPV1. Particular attention was given to the effects of numerous endogenous agonists and antagonists that can fine-tune the channel sensitivity to its usual activators, such as capsaicin, heat, acids, or their combination. In addition to the pain sensing not covered in this review, the TRPV1 channel was found to be involved in the regulation of many important physiological and pathological processes and, therefore, can be considered as a promising therapeutic target in the treatment of various diseases, such as pneumonia, ischemia, diabetes, epilepsy, schizophrenia, psoriasis, etc.
Collapse
Affiliation(s)
- Irina N Gladkikh
- Elyakov Pacific Institute of Bioorganic Chemistry, Far East Branch of the Russian Academy of Sciences, Vladivostok, 690022, Russia
| | - Oksana V Sintsova
- Elyakov Pacific Institute of Bioorganic Chemistry, Far East Branch of the Russian Academy of Sciences, Vladivostok, 690022, Russia
| | - Elena V Leychenko
- Elyakov Pacific Institute of Bioorganic Chemistry, Far East Branch of the Russian Academy of Sciences, Vladivostok, 690022, Russia
| | - Sergey A Kozlov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
| |
Collapse
|
45
|
Mitochondrial remodelling-a vicious cycle in diabetic complications. Mol Biol Rep 2021; 48:4721-4731. [PMID: 34023988 DOI: 10.1007/s11033-021-06408-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
Diabetes mellitus (DM) is a chronic, metabolic condition characterized by excessive blood glucose that causes perturbations in physiological functioning of almost all the organs of human body. This devastating metabolic disease has its implications in cognitive decline, heart damage, renal, retinal and neuronal complications that severely affects quality of life and associated with decreased life expectancy. Mitochondria possess adaptive mechanisms to meet the cellular energy demand and combat cellular stress. In recent years mitochondrial homeostasis has been point of focus where several mechanisms regulating mitochondrial health and function are evaluated. Mitochondrial dynamics plays crucial role in maintaining healthy mitochondria in cell under physiological as well as stress condition. Mitochondrial dynamics and corresponding regulating mechanisms have been implicated in progression of metabolic disorders including diabetes and its complications. In current review we have discussed about role of mitochondrial dynamics under physiological and pathological conditions. Also, modulation of mitochondrial fission and fusion in diabetic complications are described. The available literature supports mitochondrial remodelling as reliable target for diabetic complications.
Collapse
|
46
|
McEwan S, Kwon H, Tahiri A, Shanmugarajah N, Cai W, Ke J, Huang T, Belton A, Singh B, Wang L, Pang ZP, Dirice E, Engel EA, El Ouaamari A. Deconstructing the origins of sexual dimorphism in sensory modulation of pancreatic β cells. Mol Metab 2021; 53:101260. [PMID: 34023484 PMCID: PMC8258979 DOI: 10.1016/j.molmet.2021.101260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/29/2021] [Accepted: 05/17/2021] [Indexed: 01/02/2023] Open
Abstract
The regulation of glucose-stimulated insulin secretion and glucose excursion has a sensory component that operates in a sex-dependent manner. OBJECTIVE Here, we aim to dissect the basis of the sexually dimorphic interaction between sensory neurons and pancreatic β cells and its overall impact on insulin release and glucose homeostasis. METHODS We used viral retrograde tracing techniques, surgical and chemodenervation models, and primary cell-based co-culture systems to uncover the biology underlying sex differences in sensory modulation of pancreatic β-cell activity. RESULTS Retrograde transsynaptic labeling revealed a sex difference in the density of sensory innervation in the pancreas. The number of sensory neurons emanating from the dorsal root and nodose ganglia that project in the pancreas is higher in male than in female mice. Immunostaining and confocal laser scanning microscopy confirmed the higher abundance of peri-islet sensory axonal tracts in the male pancreas. Capsaicin-induced sensory chemodenervation concomitantly enhanced glucose-stimulated insulin secretion and glucose clearance in male mice. These metabolic benefits were blunted when mice were orchidectomized prior to the ablation of sensory nerves. Interestingly, orchidectomy also lowered the density of peri-islet sensory neurons. In female mice, capsaicin treatment did not affect glucose-induced insulin secretion nor glucose excursion and ovariectomy did not modify these outcomes. Interestingly, same- and opposite-sex sensory-islet co-culture paradigms unmasked the existence of potential gonadal hormone-independent mechanisms mediating the male-female difference in sensory modulation of islet β-cell activity. CONCLUSION Taken together, these data suggest that the sex-biased nature of the sensory control of islet β-cell activity is a result of a combination of neurodevelopmental inputs, sex hormone-dependent mechanisms and the potential action of somatic molecules encoded by the sex chromosome complement.
Collapse
Affiliation(s)
- Sara McEwan
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Hyokjoon Kwon
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Azeddine Tahiri
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Nivetha Shanmugarajah
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| | - Weikang Cai
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| | - Jin Ke
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Tianwen Huang
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ariana Belton
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Bhagat Singh
- F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Le Wang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Zhiping P. Pang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Ercument Dirice
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY, 10595, USA
| | - Esteban A. Engel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, 08544, 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, 08901, USA,The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA,Corresponding author. Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
47
|
Günel Z, Varhan E, Koç M, Topuz A, Sahin-Nadeem H. Production of pungency-suppressed capsaicin microcapsules by spray chilling. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.100918] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
48
|
Maruyama K. Senso-immunology: crosstalk between nociceptive and immune systems. FEBS J 2021; 289:4132-4145. [PMID: 33780155 DOI: 10.1111/febs.15846] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/03/2021] [Accepted: 03/26/2021] [Indexed: 12/21/2022]
Abstract
Understanding the molecular mechanisms of nociception has recently grown impressively. Nociception is mediated by mechanical, chemical, or microbial stimuli that evoke unpleasant feelings, alerting the host of the risk of tissue damage. Such diverse arrays of noxious stimuli trigger various escape reactions, usually altering immune homeostasis. Notably, nociceptors can recognize cytokines or pathogens via sensory molecules or innate immune receptors, participating in immune responses. Accumulating evidence suggests that activated nociceptors produce various humoral factors that affect the immune system and act like endocrine/paracrine signals. Thus, understanding the interplay between the nociceptive and immune systems may open new avenues for the development of an interdisciplinary research field, hereinafter referred to as 'senso-immunology'. This review will discuss the physiological relevance of the senso-immune system in the host defense context, focusing on how senso-immune research might yield novel treatments to cure pain and inflammation.
Collapse
Affiliation(s)
- Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| |
Collapse
|
49
|
Gautron L. The Phantom Satiation Hypothesis of Bariatric Surgery. Front Neurosci 2021; 15:626085. [PMID: 33597843 PMCID: PMC7882491 DOI: 10.3389/fnins.2021.626085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/06/2021] [Indexed: 01/26/2023] Open
Abstract
The excitation of vagal mechanoreceptors located in the stomach wall directly contributes to satiation. Thus, a loss of gastric innervation would normally be expected to result in abrogated satiation, hyperphagia, and unwanted weight gain. While Roux-en-Y-gastric bypass (RYGB) inevitably results in gastric denervation, paradoxically, bypassed subjects continue to experience satiation. Inspired by the literature in neurology on phantom limbs, I propose a new hypothesis in which damage to the stomach innervation during RYGB, including its vagal supply, leads to large-scale maladaptive changes in viscerosensory nerves and connected brain circuits. As a result, satiation may continue to arise, sometimes at exaggerated levels, even in subjects with a denervated or truncated stomach. The same maladaptive changes may also contribute to dysautonomia, unexplained pain, and new emotional responses to eating. I further revisit the metabolic benefits of bariatric surgery, with an emphasis on RYGB, in the light of this phantom satiation hypothesis.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Center for Hypothalamic Research, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
50
|
Zhang D, Yang B, Chang SQ, Ma SS, Sun JX, Yi L, Li X, Shi HM, Jing B, Zheng YC, Zhang CL, Chen FG, Zhao GP. Protective effect of paeoniflorin on H 2O 2 induced Schwann cells injury based on network pharmacology and experimental validation. Chin J Nat Med 2021; 19:90-99. [PMID: 33641788 DOI: 10.1016/s1875-5364(21)60010-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Indexed: 02/07/2023]
Abstract
This study was to investigate the protective effect of paeoniflorin (PF) on hydrogen peroxide-induced injury. Firstly, "SMILES" of PF was searched in Pubchem and further was used for reverse molecular docking in Swiss Target Prediction database to obtain potential targets. Injury-related molecules were obtained from GeenCards database, and the predicted targets of PF for injury treatment were selected by Wayne diagram. For mechanism analysis, the protein-protein interactions were constructed by String, and the KEGG analysis was conducted in Webgestalt. Then, cell viability and cytotoxicity assay were established by CCK8 assay. Also, the experimental cells were allocated to control, model (200 μmol·L-1 H2O2), SB203580 10 μmol·L-1 (200 μmol·L-1 H2O2+ SB203580 10 μmol·L-1), PF 50 μmol·L-1 (200 μmol·L-1 H2O2+ PF 50 μmol·L-1), and PF 100 μmol·L-1 (200 μmol·L-1 H2O2+ PF 100 μmol·L-1) groups. We measured the intracellular ROS, Hoechst 33258 staining, cell apoptosis, the levels of Bcl-xl, Bcl-2, Caspase-3, Cleaved-caspase3, Cleaved-caspase7, TRPA1, TRPV1, and the phosphorylation expression of p38MAPK. There are 96 potential targets that may be associated with PF for injury treatment. Then, we chose the "Inflammatory mediator regulation of TRP channels" pathway for the experimental verification from the first 10 KEGG pathway. In experimental verification, H2O2 decreased the cell viability moderately (P < 0.05), and 100 μmol·L -1 PF increased the cell viability significantly (P < 0.05). Depending on the difference of intracellular ROS fluorescence intensity, PF inhibited H 2O2-induced reactive oxygen species production in Schwann cells. In Hoechst 33258 staining, PF reversed the condensed chromatin and apoptotic nuclei following H2O2 treatment. Moreover, Flow cytometry results showed that PF could substantially inhibit H2O2 induced apoptosis (P < 0.05). Pretreatment with PF obviously reduced the levels of Caspase3, Cleaved-caspase3, Cleaved-caspase7, TRPA1, TRPV1, and the phosphorylation expression of p38MAPK after H 2O2 treatment (P < 0.05), increased the levels of Bcl-2, and Bcl-xl ( P < 0.05). PF inhibited Schwann cell injury and apoptosis induced by hydrogen peroxide, which mechanism was linked to the inhibition of phosphorylation of p38MAPK.
Collapse
Affiliation(s)
- Di Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Bing Yang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Shi-Quan Chang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Sheng-Suo Ma
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Jian-Xin Sun
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Lin Yi
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Xing Li
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Hui-Mei Shi
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Bei Jing
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Ya-Chun Zheng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Chun-Lan Zhang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China
| | - Feng-Guo Chen
- LiWan Hospital of Traditional Chinese Medicine, Guangzhou 510665, China
| | - Guo-Ping Zhao
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510630, China.
| |
Collapse
|