1
|
Zheng H, Chen Y, Lu S, Liu Z, Ma Y, Zhang C, Zhang Y, Zhang J, Liu C, Chu M, Pei F, Liu S, Duan L. Mechanosensory Piezo2 regulated by gut microbiota participates in the development of visceral hypersensitivity and intestinal dysmotility. Gut Microbes 2025; 17:2497399. [PMID: 40296251 PMCID: PMC12045567 DOI: 10.1080/19490976.2025.2497399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 01/23/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
The gut microbiota plays a crucial role in the manifestation of intestinal dysfunction associated with irritable bowel syndrome (IBS). The mechanosensory Piezo2 has been implicated in the regulation of intestinal function. However, it remains unclear whether Piezo2 is modulated by the gut microbiota, thus contributing to the development of visceral hypersensitivity and gut dysmotility. The study enrolled patients with diarrhea-predominant IBS (IBS-D) alongside healthy controls (HC). Questionnaires, rectal barostat test, and colonoscopy with mucosal biopsy were conducted. Fecal microbiota transplantation (FMT) was performed using samples from HC or IBS-D patients, and interventions with Akkermansia muciniphila or Fusobacterium varium were carried out on colon- or dorsal root ganglion (DRG)- Piezo2 knockdown pseudo-germ-free mice. Visceral sensitivity and intestinal motility were assessed. Piezo2 levels were detected using western blot and immunofluorescence. Fecal 16S rRNA sequencing and cecum untargeted metabolomics analysis, followed by molecular docking predictions of Piezo2, were also performed. The ratio of Piezo2+/5-HT+ cells was lower in IBS-D patients, positively correlated with visceral sensation and intestinal dysbiosis. The mice that received FMT from IBS-D patients exhibited colonic dysmotility and visceral hypersensitivity, along with elevated Piezo2 protein levels in the colon and DRG. Knockdown of Piezo2 in the colon or DRG ameliorated the FMT-induced colonic dysmotility and visceral hypersensitivity. Fecal 16S rRNA sequencing revealed distinct microbiota composition. Notably, Fusobacterium varium, but not Akkermansia muciniphila, induced gut dysmotility and visceral hypersensitivity, effects that could be alleviated by colon or DRG Piezo2 knockdown. Additionally, Fusobacterium varium lead to increased Piezo2 protein levels, as well as elevated levels of indole-3-acetic acid and indole-3-acrylic acid, which were predicted to bind to Piezo2, causing disturbances. Piezo2 can be regulated by gut microbiota and involved in visceral hypersensitivity and colonic dysmotility, with Fusobacterium varium playing a crucial role.
Collapse
Affiliation(s)
- Haonan Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
- PKUMed-EKEMed Joint Laboratory for Human Microbiome Research, Peking University Health Science Center, Beijing, P. R. China
| | - Yuzhu Chen
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
- PKUMed-EKEMed Joint Laboratory for Human Microbiome Research, Peking University Health Science Center, Beijing, P. R. China
| | - Siqi Lu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
| | - Zuojing Liu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
| | - Yinchao Ma
- Department of Immunology, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China
| | - Cunzheng Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
- PKUMed-EKEMed Joint Laboratory for Human Microbiome Research, Peking University Health Science Center, Beijing, P. R. China
| | - Yiming Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
| | - Jindong Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
- PKUMed-EKEMed Joint Laboratory for Human Microbiome Research, Peking University Health Science Center, Beijing, P. R. China
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Ming Chu
- Department of Immunology, NHC Key Laboratory of Medical Immunology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China
| | - Fei Pei
- Department of Pathology, Peking University Third Hospital; School of Basic Medical Sciences, Peking University, Beijing, P. R. China
| | - Shuangjiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing, P. R. China
- Beijing Key Laboratory for Helicobacter pylori Infection and Upper Gastrointestinal Diseases, Peking University Third Hospital, Beijing, P. R. China
- PKUMed-EKEMed Joint Laboratory for Human Microbiome Research, Peking University Health Science Center, Beijing, P. R. China
| |
Collapse
|
2
|
Xie Z, Rose L, Feng J, Zhao Y, Lu Y, Kane H, Hibberd TJ, Hu X, Wang Z, Zang K, Yang X, Richardson Q, Othman R, Venezia O, Zhakyp A, Gao F, Abe N, Vigeland K, Wang H, Branch C, Duizer C, Deng L, Meng X, Zamidar L, Hauptschein M, Bergin R, Dong X, Chiu IM, Kim BS, Spencer NJ, Hu H, Jackson R. Enteric neuronal Piezo1 maintains mechanical and immunological homeostasis by sensing force. Cell 2025; 188:2417-2432.e19. [PMID: 40132579 PMCID: PMC12048284 DOI: 10.1016/j.cell.2025.02.031] [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: 05/06/2024] [Revised: 11/19/2024] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
The gastrointestinal (GI) tract experiences a myriad of mechanical forces while orchestrating digestion and barrier immunity. A central conductor of these processes, the enteric nervous system (ENS), detects luminal pressure to regulate peristalsis independently of extrinsic input from the central and peripheral nervous systems. However, how the ∼500 million enteric neurons that reside in the GI tract sense and respond to force remains unknown. Herein, we establish that the mechanosensor Piezo1 is functionally expressed in cholinergic enteric neurons. Optogenetic stimulation of Piezo1+ cholinergic enteric neurons drives colonic motility, while Piezo1 deficiency reduces cholinergic neuronal activity and slows peristalsis. Additionally, Piezo1 deficiency in cholinergic enteric neurons abolishes exercise-induced acceleration of GI motility. Finally, we uncover that enteric neuronal Piezo1 function is required for motility alterations in colitis and acts to prevent aberrant inflammation and tissue damage. This work uncovers how the ENS senses and responds to mechanical force.
Collapse
Affiliation(s)
- Zili Xie
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Lillian Rose
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghui Zhao
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Yisi Lu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Harry Kane
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Xueming Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Zhen Wang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Kaikai Zang
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Xingliang Yang
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | | | - Rahmeh Othman
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Olivia Venezia
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ademi Zhakyp
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Fang Gao
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Nobuya Abe
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keren Vigeland
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Hongshen Wang
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Camren Branch
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Coco Duizer
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Xia Meng
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Lydia Zamidar
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Max Hauptschein
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ronan Bergin
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Issac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Brian S Kim
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Hongzhen Hu
- Department of Dermatology, The Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO 63130, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, New York, NY 10029, USA; The Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ruaidhrí Jackson
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Abdullah NS, Bradaia A, Defaye M, Ohland C, Svendsen K, Dickemann A, Delanne-Cumenal M, Hassan A, Iftinca M, McCoy KD, Altier C. Early life microbiota colonization programs nociceptor sensitivity by regulating NGF production in mast cells. Mucosal Immunol 2025; 18:326-338. [PMID: 39662673 DOI: 10.1016/j.mucimm.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Recent evidence suggests that the gut microbiota can influence pain sensitivity, highlighting the potential for microbiota-targeted pain interventions. During early life, both the microbiota and nociceptors are fine-tuned and respond to environmental factors, however, little is known about how they interact with each other. Using germ-free and gnotobiotic models, we demonstrate that microbiota colonization controls nociceptor sensitivity, partly by modulating mast cell production of nerve growth factor (NGF). We report that germ-free mice respond less to thermal and capsaicin-induced stimulation, which correlates with reduced trafficking of TRPV1 to the cell membrane of nociceptors. In germ-free mice, mast cells express lower levels of NGF. Hyposensitivity to thermal and capsaicin-induced stimulation, reduced TRPV1 trafficking, and decreased NGF expression are reversed when mice are colonized at birth, but not when colonization occurs after weaning. Inhibition of mast cell degranulation and NGF signaling during the first weeks of life in colonized mice leads to a hyposensitive phenotype in adulthood, demonstrating a role for mast cells and NGF signaling in linking early life colonization with nociceptor sensitivity. These findings implicate the early life microbiota in shaping mast cell NGF production and nociceptor sensitivity later in life. SIGNIFICANCE STATEMENT: Nociceptors are specialized sensory neurons that detect and transduce painful stimuli. During the early postnatal period, nociceptors are influenced by sensory experiences and the environment. Our findings demonstrate that gut microbiota colonization is essential in setting the threshold of nociceptor responses to painful stimuli. We show that early-life bacterial colonization controls the production of nerve growth factor by mast cells, affecting our sensitivity to pain later in life. Our study highlights the potential for developing new pain treatments that target the gut microbiome.
Collapse
Affiliation(s)
- Nasser S Abdullah
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Amyaouch Bradaia
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Manon Defaye
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Christina Ohland
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Kristofer Svendsen
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Anabel Dickemann
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Melissa Delanne-Cumenal
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Ahmed Hassan
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Mircea Iftinca
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Christophe Altier
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Inflammation Research Network, Snyder institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, T2N4N1, Canada.
| |
Collapse
|
4
|
Xie W, Lückemeyer DD, Qualls KA, Prudente AS, Berta T, Gu M, Strong JA, Dong X, Zhang JM. Vascular motion in the dorsal root ganglion sensed by Piezo2 in sensory neurons triggers episodic neuropathic pain. Neuron 2025:S0896-6273(25)00178-3. [PMID: 40154477 DOI: 10.1016/j.neuron.2025.03.006] [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: 09/12/2024] [Revised: 01/27/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Spontaneous pain, characterized by episodic shooting or stabbing sensations, is a major complaint among neuropathic pain patients, yet its mechanisms remain poorly understood. Recent research indicates a connection between this pain condition and "clustered firing," wherein adjacent sensory neurons fire simultaneously. This study presents evidence that the triggers of spontaneous pain and clustered firing are the dynamic movements of small blood vessels within the nerve-injured sensory ganglion, along with increased blood vessel density/angiogenesis and increased number of pericytes around blood vessels. Pharmacologically or mechanically evoked myogenic vascular responses increase both spontaneous pain and clustered firing in a mouse model of neuropathic pain. The mechanoreceptor Piezo2 in sensory neurons plays a critical role in detecting blood vessel movements. An anti-VEGF monoclonal antibody that inhibits angiogenesis effectively blocks spontaneous pain and clustered firing. These findings suggest targeting Piezo2, angiogenesis, or abnormal vascular dynamics as potential therapeutic strategies for neuropathic spontaneous pain.
Collapse
Affiliation(s)
- Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Debora Denardin Lückemeyer
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Katherine A Qualls
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mingxia Gu
- Department of Anesthesiology & Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA; Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21209, USA.
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
5
|
Luo J, Xu Q, Xu S, Zhai L, Yuan CS, Bian Z. Decoding Abdominal Pain in Constipation-predominant Irritable Bowel Syndrome and Functional Constipation: Mechanisms and Managements. Curr Gastroenterol Rep 2025; 27:22. [PMID: 40095229 PMCID: PMC11914341 DOI: 10.1007/s11894-025-00967-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW Abdominal pain in constipation-predominant irritable bowel syndrome (IBS-C) and functional constipation (FC) remains a difficult clinical challenge due to unclear pathophysiological mechanisms and limited pain-targeted treatments. This review critically evaluates the evidence on the underlying pain mechanisms in IBS-C and/or FC and explores management strategies, their limitations, and future directions. RECENT FINDINGS Most research on constipation-related pain is based on IBS-C patients or animal models, with limited studies focusing on FC. Visceral hypersensitivity, serotonin dysregulation, gut-brain axis dysfunction, and central/peripheral nervous system alterations are implicated in IBS-C pain, while FC pain is less studied and may be primarily linked to colonic distension and motility dysfunction. Management strategies include 5-HT4 agonists, GC-C agonists, chloride channel activators, psychological therapies, probiotics and complementary medicine. Despite available treatment options, managing abdominal pain in IBS-C and FC remains challenging due to heterogeneous pathophysiology and limited targeted therapies. While some interventions provide symptomatic relief, there is no universally effective treatment for abdominal pain across all patients. Future research should focus on identifying pain-specific biomarkers, refining diagnostic criteria, and integrating multi-omics data and neuroimaging techniques to better distinguish pain mechanisms in IBS-C versus FC and develop more precise, patient-centered interventions.
Collapse
Affiliation(s)
- Jingyuan Luo
- Vincent V.C. Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, 3/F, Jockey Club School of Chinese Medicine Building, 7 Baptist University Road, Kowloon Tong, Hong Kong, SAR, China
- Center for Chinese Herbal Medicine Drug Development and School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Qianqian Xu
- Vincent V.C. Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, 3/F, Jockey Club School of Chinese Medicine Building, 7 Baptist University Road, Kowloon Tong, Hong Kong, SAR, China
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14214-8033, USA
| | - Shujun Xu
- Center for Chinese Herbal Medicine Drug Development and School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Lixiang Zhai
- Center for Chinese Herbal Medicine Drug Development and School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research and Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, 5841 South Maryland Avenue, MC 4028, Chicago, IL, 60637, USA.
- Department of Anesthesia and Critical Care, Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA.
| | - Zhaoxiang Bian
- Vincent V.C. Woo Chinese Medicine Clinical Research Institute, School of Chinese Medicine, Hong Kong Baptist University, 3/F, Jockey Club School of Chinese Medicine Building, 7 Baptist University Road, Kowloon Tong, Hong Kong, SAR, China.
- Center for Chinese Herbal Medicine Drug Development and School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, China.
| |
Collapse
|
6
|
Quijas MM, Queme LF, Woodke ST, Weyler AA, Buesing D, Butterfield A, Joshi DP, Mitxelena I, Ulrich-Lai YM, Jankowski MP. Sex-specific role of RNA-binding protein, pAUF1, on prolonged hypersensitivity after repetitive ischemia with reperfusion injury. Pain 2025; 166:693-707. [PMID: 39382317 PMCID: PMC11810611 DOI: 10.1097/j.pain.0000000000003415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT Repetitive ischemia with reperfusion (I/R) injury is a common cause of myalgia. Ischemia with reperfusion injuries occur in many conditions that differentially affect males and females including complex regional pain syndrome and fibromyalgia. Our preclinical studies have indicated that primary afferent sensitization and behavioral hypersensitivity caused by I/R injury may be due to sex-specific gene expression in the dorsal root ganglia (DRG) and distinct upregulation of growth factors and cytokines in the affected muscles. To determine how these unique gene expression programs may be established in a sex-dependent manner in a model that more closely mimics clinical scenarios, we used a developed prolonged ischemic myalgia model in mice whereby animals experience repeated I/R injuries and compared behavioral results with unbiased and targeted screening strategies in male and female DRG. Several distinct proteins were found to be differentially expressed in male and female DRG, including phosphorylated AU-rich element RNA-binding protein (pAUF1), which is known to regulate gene expression. Nerve-specific siRNA-mediated knockdown of AUF1 inhibited prolonged hypersensitivity in females only, whereas overexpression of AUF1 in male DRG neurons increased pain-like responses. AUF1 knockdown was able to specifically inhibit repeated I/R-induced gene expression in females potentially downstream of prolactin receptor signaling. Data suggest RNA-binding proteins such as pAUF1 may underlie the sex-specific effects on DRG gene expression that modulates behavioral hypersensitivity after repeated I/R injury through prolactin signaling. This study may aid in finding distinct receptor differences related to the evolution of acute to chronic ischemic muscle pain development between sexes.
Collapse
Affiliation(s)
- Meranda M. Quijas
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Luis F. Queme
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Samantha T. Woodke
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Alex A. Weyler
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Dana Buesing
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Ally Butterfield
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Diya P. Joshi
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Irati Mitxelena
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Yvonne M. Ulrich-Lai
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Michael P. Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Pediatric Pain Research Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
7
|
Wolfson RL. Spinal sensory innervation of the intestine. Curr Opin Neurobiol 2025; 90:102973. [PMID: 39892315 PMCID: PMC11951475 DOI: 10.1016/j.conb.2025.102973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/31/2024] [Accepted: 01/07/2025] [Indexed: 02/03/2025]
Abstract
Sensing our internal environment, or interoception, is essential under physiologic circumstances, such as controlling food intake, and under pathophysiologic circumstances, often triggering abdominal pain. The sensory neurons that innervate the gastrointestinal (GI) tract to mediate interoception originate in two separate parts of the peripheral nervous system: the spinal sensory neurons, whose cell bodies reside in the dorsal root ganglia (DRG), and the vagal sensory neurons, whose cell bodies reside in the nodose ganglia. While the vagal sensory neurons have been extensively studied for their roles in interoception, the roles of the DRG sensory neurons in internal gut sensing are only beginning to be uncovered. Here, we review the recent advances in understanding the diverse properties and functions of gut-innervating DRG sensory neurons and highlight the many unknowns with regards to this understudied population in regulating interoception.
Collapse
Affiliation(s)
- Rachel L Wolfson
- Department of Cell Biology, Harvard Medical School, 250 Longwood Avenue, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
8
|
Wan Y, Zhou J, Li H. The Role of Mechanosensitive Piezo Channels in Chronic Pain. J Pain Res 2024; 17:4199-4212. [PMID: 39679432 PMCID: PMC11646438 DOI: 10.2147/jpr.s490459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose of Review Mechanosensitive Piezo channels are ion channels activated by mechanical stimuli, playing a crucial role in mechanotransduction processes and mechanical hypersensitivity. When these channels are subjected to mechanical loading, membrane currents rise instantaneously, depolarizing and activating voltage-gated calcium channels. This results in an increase in intracellular Ca2+, which contributes to heightened sensitivity to mechanical stimuli. This review delves into the characteristics and mechanisms of Piezo channels in chronic pain. Recent Findings The findings suggest that Piezo channels are integral to the occurrence and development of chronic pain, including neuropathic pain, visceral pain, musculoskeletal pain, headache or orofacial pain, and inflammatory pain. Piezo channels significantly impact pain perception and transmission. These channels' critical involvement in various pain types highlights their potential as promising targets for chronic pain therapy. Summary This review discusses the role of Piezo channels in chronic pain. By understanding these pain mechanisms, new therapeutic strategies can be developed to alleviate chronic pain, offering hope for patients suffering from these debilitating conditions.
Collapse
Affiliation(s)
- Yantong Wan
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, People’s Republic of China
| | - Jieshu Zhou
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, People’s Republic of China
| | - Hao Li
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
9
|
Wang Z, Song K, Kim BS, Manion J. Sensory neuroimmune interactions at the barrier. Mucosal Immunol 2024; 17:1151-1160. [PMID: 39374664 DOI: 10.1016/j.mucimm.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Epithelial barriers such as the skin, lung, and gut, in addition to having unique physiologic functions, are designed to preserve tissue homeostasis upon challenge with a variety of allergens, irritants, or pathogens. Both the innate and adaptive immune systems play a critical role in responding to epithelial cues triggered by environmental stimuli. However, the mechanisms by which organs sense and coordinate complex epithelial, stromal, and immune responses have remained a mystery. Our increasing understanding of the anatomic and functional characteristics of the sensory nervous system is greatly advancing a new field of peripheral neuroimmunology and subsequently changing our understanding of mucosal immunology. Herein, we detail how sensory biology is informing mucosal neuroimmunology, even beyond neuroimmune interactions seen within the central and autonomic nervous systems.
Collapse
Affiliation(s)
- Zhen Wang
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
| | - Keaton Song
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian S Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA.
| | - John Manion
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Urology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Surgery, Harvard Medical School, Boston, MA 02115, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Song K, Kim BS. The peripheral neuroimmune system. J Leukoc Biol 2024; 116:1291-1300. [PMID: 39422243 PMCID: PMC11599120 DOI: 10.1093/jleuko/qiae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/18/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024] Open
Abstract
Historically, the nervous and immune systems were studied as separate entities. The nervous system relays signals between the body and the brain by processing sensory inputs and executing motor outputs, whereas the immune system provides protection against injury and infection through inflammation. However, recent developments have demonstrated that these systems mount tightly integrated responses. In particular, the peripheral nervous system acts in concert with the immune system to control reflexes that maintain and restore homeostasis. Notwithstanding their homeostatic mechanisms, dysregulation of these neuroimmune interactions may underlie various pathological conditions. Understanding how these two distinct systems communicate is an emerging field of peripheral neuroimmunology that promises to reveal new insights into tissue physiology and identify novel targets to treat disease.
Collapse
Affiliation(s)
- Keaton Song
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
| | - Brian S Kim
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai, 787 11th Ave, New York, NY 10019, USA
| |
Collapse
|
11
|
Li YC, Zhang FC, Li D, Weng RX, Yu Y, Gao R, Xu GY. Distinct circuits and molecular targets of the paraventricular hypothalamus decode visceral and somatic pain. Neuron 2024; 112:3734-3749.e5. [PMID: 39326407 DOI: 10.1016/j.neuron.2024.08.024] [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: 03/27/2024] [Revised: 07/17/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
Visceral and somatic pain serve as protective mechanisms against external threats. Accumulated evidence has confirmed that the paraventricular hypothalamus (PVH) plays an important role in the perception of visceral and somatic pain, whereas the exact neural pathways and molecules distinguishing them remain unclear. Here, we report distinct neuronal ensembles within the PVH dedicated to processing visceral and somatic pain signals. An essential discovery is the distinct expression of P2X3R and VIPR2 in visceral and somatic pain-activated PVH neuronal ensembles. Furthermore, visceral pain- and somatic pain-responsive PVH neuronal ensembles project to specific downstream regions, the ventral part of the lateral septal nucleus (LSV) and the caudal part of the zona incerta (ZIC), respectively. These findings unveil that the PVH acts as a pain sorting center that distinctly processes visceral and somatic pain, identifying potential molecular targets for specific pain processing and providing a new framework for comprehending how the brain processes nociceptive information.
Collapse
Affiliation(s)
- Yong-Chang Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China; Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Fu-Chao Zhang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Di Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Rui-Xia Weng
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China; Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, P.R. China
| | - Yang Yu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China
| | - Rong Gao
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215123, Jiangsu, P.R. China.
| | - Guang-Yin Xu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, Jiangsu, P.R. China.
| |
Collapse
|
12
|
Xiao B. Mechanisms of mechanotransduction and physiological roles of PIEZO channels. Nat Rev Mol Cell Biol 2024; 25:886-903. [PMID: 39251883 DOI: 10.1038/s41580-024-00773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Mechanical force is an essential physical element that contributes to the formation and function of life. The discovery of the evolutionarily conserved PIEZO family, including PIEZO1 and PIEZO2 in mammals, as bona fide mechanically activated cation channels has transformed our understanding of how mechanical forces are sensed and transduced into biological activities. In this Review, I discuss recent structure-function studies that have illustrated how PIEZO1 and PIEZO2 adopt their unique structural design and curvature-based gating dynamics, enabling their function as dedicated mechanotransduction channels with high mechanosensitivity and selective cation conductivity. I also discuss our current understanding of the physiological and pathophysiological roles mediated by PIEZO channels, including PIEZO1-dependent regulation of development and functional homeostasis and PIEZO2-dominated mechanosensation of touch, tactile pain, proprioception and interoception of mechanical states of internal organs. Despite the remarkable progress in PIEZO research, this Review also highlights outstanding questions in the field.
Collapse
Affiliation(s)
- Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center of Biological Structure, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
13
|
Mazor Y, Engelmayer N, Nashashibi H, Rottenfußer L, Lev S, Binshtok AM. Attenuation of Colitis-Induced Visceral Hypersensitivity and Pain by Selective Silencing of TRPV1-Expressing Fibers in Rat Colon. Inflamm Bowel Dis 2024; 30:1843-1851. [PMID: 38478397 PMCID: PMC11447070 DOI: 10.1093/ibd/izae036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Indexed: 10/04/2024]
Abstract
BACKGROUND Transient receptor potential vanilloid 1 (TRPV1) cation channels, expressed on nociceptors, are well established as key contributors to abdominal pain in inflammatory bowel disease (IBD). Previous attempts at blocking these channels have been riddled with side effects. Here, we propose a novel treatment strategy, utilizing the large pore of TRPV1 channels as a drug delivery system to selectively inhibit visceral nociceptors. METHODS We induced colitis in rats using intrarectal dinitrobenzene sulfonic acid. Visceral hypersensitivity, spontaneous pain, and responsiveness of the hind paws to noxious heat stimuli were examined before and after the intrarectal application of membrane-impermeable sodium channel blocker (QX-314) alone or together with TRPV1 channel activators or blockers. RESULTS Intrarectal co-application of QX-314 with TRPV1 channel activator capsaicin significantly inhibited colitis-induced gut hypersensitivity. Furthermore, in the model of colitis, but not in naïve rats, QX-314 alone was sufficient to reverse gut hypersensitivity. The blockade of TRPV1 channels prevented this effect of QX-314. Finally, applying QX-314 alone to the inflamed gut inhibited colitis-induced ongoing pain. CONCLUSIONS Selective silencing of gut nociceptors by a membrane-impermeable sodium channel blocker entering via exogenously or endogenously activated TRPV1 channels diminishes IBD-induced gut hypersensitivity. The lack of effect on naïve rats suggests a selective analgesic effect in the inflamed gut. Our results suggest that in the colitis model, TRPV1 channels are tonically active. Furthermore, our results emphasize the role of TRPV1-expressing nociceptive fibers in colitis-induced pain. These findings provide proof of concept for using charged activity blockers for the blockade of IBD-associated abdominal pain.
Collapse
Affiliation(s)
- Yoav Mazor
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hadassah School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
| | - Nurit Engelmayer
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hadassah School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Halla Nashashibi
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hadassah School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lisa Rottenfußer
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hadassah School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
| | - Shaya Lev
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hadassah School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alexander M Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hadassah School of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
14
|
Hamed YMF, Ghosh B, Marshall KL. PIEZO ion channels: force sensors of the interoceptive nervous system. J Physiol 2024; 602:4777-4788. [PMID: 38456626 PMCID: PMC11845038 DOI: 10.1113/jp284077] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/08/2024] [Indexed: 03/09/2024] Open
Abstract
Many organs are designed to move: the heart pumps each second, the gastrointestinal tract squeezes and churns to digest food, and we contract and relax skeletal muscles to move our bodies. Sensory neurons of the peripheral nervous system detect signals from bodily tissues, including the forces generated by these movements, to control physiology. The processing of these internal signals is called interoception, but this is a broad term that includes a wide variety of both chemical and mechanical sensory processes. Mechanical senses are understudied, but rapid progress has been made in the last decade, thanks in part to the discovery of the mechanosensory PIEZO ion channels (Coste et al., 2010). The role of these mechanosensors within the interoceptive nervous system is the focus of this review. In defining the transduction molecules that govern mechanical interoception, we will have a better grasp of how these signals drive physiology.
Collapse
Affiliation(s)
- Yasmeen M. F. Hamed
- Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Britya Ghosh
- Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kara L. Marshall
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
- Lead contact
| |
Collapse
|
15
|
Liu R, Luo Y, Ma J, Zhang Q, Sheng Y, Li J, Li H, Zhao T. Traditional Chinese medicine for functional gastrointestinal disorders and inflammatory bowel disease: narrative review of the evidence and potential mechanisms involving the brain-gut axis. Front Pharmacol 2024; 15:1444922. [PMID: 39355776 PMCID: PMC11443704 DOI: 10.3389/fphar.2024.1444922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/23/2024] [Indexed: 10/03/2024] Open
Abstract
Functional gastrointestinal disorders (FGIDs) and inflammatory bowel disease (IBD) are common clinical disorders characterized by recurrent diarrhea and abdominal pain. Although their pathogenesis has not been fully clarified, disruptions in intestinal motility and immune function are widely accepted as contributing factors to both conditions, and the brain-gut axis plays a key role in these processes. Traditional Chinese Medicine (TCM) employs a holistic approach to treatment, considers spleen and stomach impairments and liver abnormality the main pathogenesis of these two diseases, and offers a unique therapeutic strategy that targets these interconnected pathways. Clinical evidence shows the great potential of TCM in treating FGIDs and IBD. This study presents a systematic description of the pathological mechanisms of FGIDs and IBD in the context of the brain-gut axis, discusses clinical and preclinical studies on TCM and acupuncture for the treatment of these diseases, and summarizes TCM targets and pathways for the treatment of FGIDs and IBD, integrating ancient wisdom with contemporary biomedical insights. The alleviating effects of TCM on FGID and IBD symptoms are mainly mediated through the modulation of intestinal immunity and inflammation, sensory transmission, neuroendocrine-immune network, and microbiota and their metabolism through brain-gut axis mechanisms. TCM may be a promising treatment option in controlling FGIDs and IBD; however, further high-quality research is required. This review provides a reference for an in-depth exploration of the interventional effects and mechanisms of TCM in FGIDs and IBD, underscoring TCM's potential to recalibrate the dysregulated brain-gut axis in FGIDs and IBD.
Collapse
Affiliation(s)
- RuiXuan Liu
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - YunTian Luo
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - JinYing Ma
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qi Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yudong Sheng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiashan Li
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongjiao Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - TianYi Zhao
- School of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
16
|
Xia B, Lin T, Li Z, Wang J, Sun Y, Wang D, Ye J, Zhang Y, Kou R, Zhao B, Yi J, Bai G, Liu X. Lactiplantibacillus plantarum Regulates Intestinal Physiology and Enteric Neurons in IBS through Microbial Tryptophan Metabolites. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:17989-18002. [PMID: 39082086 DOI: 10.1021/acs.jafc.4c03087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder characterized by visceral pain and gut dysmotility. However, the specific mechanisms by which Lactobacillus strains relieve IBS remain unclear. Here, we screened Lactobacillus strains from traditional Chinese fermented foods with potential IBS-alleviating properties through in vitro and in vivo experiments. We demonstrated that Lactiplantibacillus plantarum D266 (Lp D266) administration effectively modulates intestinal peristalsis, enteric neurons, visceral hypersensitivity, colonic inflammation, gut barrier function, and mast cell activation. Additionally, Lp D266 shapes gut microbiota and enhances tryptophan (Trp) metabolism, thus activating the aryl hydrocarbon receptor (AhR) and subsequently enhancing IL-22 production to maintain gut homeostasis. Mechanistically, Lp D266 potentially modulates colonic physiology and enteric neurons by microbial tryptophan metabolites. Further, our study indicates that combining Lp D266 with Trp synergistically ameliorates IBS symptoms. Together, our experiments identify the therapeutic efficacy of tryptophan-catabolizing Lp D266 in regulating gut physiology and enteric neurons, providing new insights into the development of probiotic-mediated nutritional intervention for IBS management.
Collapse
Affiliation(s)
- Bing Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Tongkui Lin
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Zhiqing Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Jialin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Yuwei Sun
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Danna Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Jin Ye
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Yajuan Zhang
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Rongwei Kou
- School of Science, Xi'an University of Technology, Xi'an, 710048 Shaanxi, China
| | - Beita Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| | - Junjie Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500 Yunnan, China
| | - Gaiyan Bai
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, 710068 Shaanxi, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, 712100 Shaanxi, China
| |
Collapse
|
17
|
Guo J, Li L, Chen F, Fu M, Cheng C, Wang M, Hu J, Pei L, Sun J. Forces Bless You: Mechanosensitive Piezo Channels in Gastrointestinal Physiology and Pathology. Biomolecules 2024; 14:804. [PMID: 39062518 PMCID: PMC11274378 DOI: 10.3390/biom14070804] [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: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
The gastrointestinal (GI) tract is an organ actively involved in mechanical processes, where it detects forces via a mechanosensation mechanism. Mechanosensation relies on specialized cells termed mechanoreceptors, which convert mechanical forces into electrochemical signals via mechanosensors. The mechanosensitive Piezo1 and Piezo2 are widely expressed in various mechanosensitive cells that respond to GI mechanical forces by altering transmembrane ionic currents, such as epithelial cells, enterochromaffin cells, and intrinsic and extrinsic enteric neurons. This review highlights recent research advances on mechanosensitive Piezo channels in GI physiology and pathology. Specifically, the latest insights on the role of Piezo channels in the intestinal barrier, GI motility, and intestinal mechanosensation are summarized. Additionally, an overview of Piezo channels in the pathogenesis of GI disorders, including irritable bowel syndrome, inflammatory bowel disease, and GI cancers, is provided. Overall, the presence of mechanosensitive Piezo channels offers a promising new perspective for the treatment of various GI disorders.
Collapse
Affiliation(s)
- Jing Guo
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Li Li
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Feiyi Chen
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Minhan Fu
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Cheng Cheng
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Meizi Wang
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Jun Hu
- Health and Rehabilitation College, Nanjing University of Chinese Medicine, Nanjing 210023, China; (J.G.); (C.C.); (M.W.); (J.H.)
| | - Lixia Pei
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| | - Jianhua Sun
- Department of Acupuncture and Rehabilitation, The Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210029, China; (L.L.); (F.C.); (M.F.)
| |
Collapse
|
18
|
Cao B, Xu Q, Shi Y, Zhao R, Li H, Zheng J, Liu F, Wan Y, Wei B. Pathology of pain and its implications for therapeutic interventions. Signal Transduct Target Ther 2024; 9:155. [PMID: 38851750 PMCID: PMC11162504 DOI: 10.1038/s41392-024-01845-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/10/2024] Open
Abstract
Pain is estimated to affect more than 20% of the global population, imposing incalculable health and economic burdens. Effective pain management is crucial for individuals suffering from pain. However, the current methods for pain assessment and treatment fall short of clinical needs. Benefiting from advances in neuroscience and biotechnology, the neuronal circuits and molecular mechanisms critically involved in pain modulation have been elucidated. These research achievements have incited progress in identifying new diagnostic and therapeutic targets. In this review, we first introduce fundamental knowledge about pain, setting the stage for the subsequent contents. The review next delves into the molecular mechanisms underlying pain disorders, including gene mutation, epigenetic modification, posttranslational modification, inflammasome, signaling pathways and microbiota. To better present a comprehensive view of pain research, two prominent issues, sexual dimorphism and pain comorbidities, are discussed in detail based on current findings. The status quo of pain evaluation and manipulation is summarized. A series of improved and innovative pain management strategies, such as gene therapy, monoclonal antibody, brain-computer interface and microbial intervention, are making strides towards clinical application. We highlight existing limitations and future directions for enhancing the quality of preclinical and clinical research. Efforts to decipher the complexities of pain pathology will be instrumental in translating scientific discoveries into clinical practice, thereby improving pain management from bench to bedside.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qixuan Xu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Yajiao Shi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
19
|
Ford AC, Vanner S, Kashyap PC, Nasser Y. Chronic Visceral Pain: New Peripheral Mechanistic Insights and Resulting Treatments. Gastroenterology 2024; 166:976-994. [PMID: 38325759 PMCID: PMC11102851 DOI: 10.1053/j.gastro.2024.01.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/15/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
Chronic visceral pain is one of the most common reasons for patients with gastrointestinal disorders, such as inflammatory bowel disease or disorders of brain-gut interaction, to seek medical attention. It represents a substantial burden to patients and is associated with anxiety, depression, reductions in quality of life, and impaired social functioning, as well as increased direct and indirect health care costs to society. Unfortunately, the diagnosis and treatment of chronic visceral pain is difficult, in part because our understanding of the underlying pathophysiologic basis is incomplete. In this review, we highlight recent advances in peripheral pain signaling and specific physiologic and pathophysiologic preclinical mechanisms that result in the sensitization of peripheral pain pathways. We focus on preclinical mechanisms that have been translated into treatment approaches and summarize the current evidence base for directing treatment toward these mechanisms of chronic visceral pain derived from clinical trials. The effective management of chronic visceral pain remains of critical importance for the quality of life of suffers. A deeper understanding of peripheral pain mechanisms is necessary and may provide the basis for novel therapeutic interventions.
Collapse
Affiliation(s)
- Alexander C Ford
- Leeds Institute of Medical Research at St. James's, University of |Leeds, Leeds, United Kingdom; Leeds Gastroenterology Institute, Leeds Teaching Hospitals National Health Service Trust, Leeds, United Kingdom
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario, Canada
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
20
|
Tiwari N, Smith C, Sharma D, Shen S, Mehta P, Qiao LY. Plp1-expresssing perineuronal DRG cells facilitate colonic and somatic chronic mechanical pain involving Piezo2 upregulation in DRG neurons. Cell Rep 2024; 43:114230. [PMID: 38743566 PMCID: PMC11234328 DOI: 10.1016/j.celrep.2024.114230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/06/2024] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
Satellite glial cells (SGCs) of dorsal root ganglia (DRGs) are activated in a variety of chronic pain conditions; however, their mediation roles in pain remain elusive. Here, we take advantage of proteolipid protein (PLP)/creERT-driven recombination in the periphery mainly occurring in SGCs of DRGs to assess the role of SGCs in the regulation of chronic mechanical hypersensitivity and pain-like responses in two organs, the distal colon and hindpaw, to test generality. We show that PLP/creERT-driven hM3Dq activation increases, and PLP/creERT-driven TrkB.T1 deletion attenuates, colon and hindpaw chronic mechanical hypersensitivity, positively associating with calcitonin gene-related peptide (CGRP) expression in DRGs and phospho-cAMP response element-binding protein (CREB) expression in the dorsal horn of the spinal cord. Activation of Plp1+ DRG cells also increases the number of small DRG neurons expressing Piezo2 and acquiring mechanosensitivity and leads to peripheral organ neurogenic inflammation. These findings unravel a role and mechanism of Plp1+ cells, mainly SGCs, in the facilitation of chronic mechanical pain and suggest therapeutic targets for pain mitigation.
Collapse
Affiliation(s)
- Namrata Tiwari
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Cristina Smith
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Divya Sharma
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Shanwei Shen
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Parshva Mehta
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA
| | - Liya Y Qiao
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298-0551, USA.
| |
Collapse
|
21
|
Yang W, Lin L, Hu S, Jiang B, Yang R, Yu W, Tang J, Zhao D, Gu Y, Jin M, Li J, Lu E. Expression patterns of mechanosensitive ion channel PIEZOs in irreversible pulpitis. BMC Oral Health 2024; 24:465. [PMID: 38627713 PMCID: PMC11022356 DOI: 10.1186/s12903-024-04209-6] [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: 01/24/2024] [Accepted: 03/30/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Mechanosensitive ion channel PIEZOs have been widely reported to involve inflammation and pain. This study aimed to clarify expression patterns of PIEZOs and their potential relations to irreversible pulpitis. MATERIALS AND METHODS Normal pulp tissues (n = 29) from patients with impacted third molars and inflamed pulp tissues (n = 23) from patients with irreversible pulpitis were collected. Pain levels were assessed using a numerical rating scale. PIEZO expressions were measured using real-time PCR and then confirmed using GEO datasets GSE77459, immunoblot, and immunohistochemistry staining. Correlations of PIEZO mRNA expression with inflammatory markers, pain markers, or clinical pain levels were evaluated using Spearman's correlation analysis. Univariate analysis was conducted to analyze PIEZO expressions based on pain description and clinical examinations of cold test, percussion, palpation, and bite test. RESULTS Compared with normal pulp tissues, mRNA expression levels of PIEZO1 were significantly increased in inflamed pulp tissues, while PIEZO2 was significantly decreased, which was further confirmed in GSE77459 and on a protein and histological level. The positive correlation of the mRNA expression levels between PIEZO1 and inflammatory markers, as well as between PIEZO2 and pain markers, was verified. PIEZO2 expression was also positively correlated with pain levels. Besides, irreversible pulpitis patients who reported continuous pain and who detected a positive response to cold stimulus exhibited a higher expression level of PIEZO2 in the inflamed pulp tissues. By contrast, patients reporting pain duration of more than one week showed a higher expression level of PIEZO1. CONCLUSIONS This study demonstrated the upregulation of PIEZO1 and the downregulation of PIEZO2 in irreversible pulpitis and revealed the potential relation of PIEZO1 and PIEZO2 to inflammation and pain. These findings suggested that PIEZOs might play critical roles in the progression of irreversible pulpitis and paved the way for further investigations aimed at novel therapies of irreversible pulpitis by targeting PIEZOs.
Collapse
Affiliation(s)
- Wenying Yang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Lu Lin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Shucheng Hu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Bin Jiang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ruhan Yang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Weijun Yu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Jiaqi Tang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Dan Zhao
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yuting Gu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China.
| | - Min Jin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China.
| | - Jin Li
- Department of Ophthalmology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China.
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
22
|
Gupta S, Viotti A, Eichwald T, Roger A, Kaufmann E, Othman R, Ghasemlou N, Rafei M, Foster SL, Talbot S. Navigating the blurred path of mixed neuroimmune signaling. J Allergy Clin Immunol 2024; 153:924-938. [PMID: 38373475 DOI: 10.1016/j.jaci.2024.02.006] [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: 10/11/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/21/2024]
Abstract
Evolution has created complex mechanisms to sense environmental danger and protect tissues, with the nervous and immune systems playing pivotal roles. These systems work together, coordinating local and systemic reflexes to restore homeostasis in response to tissue injury and infection. By sharing receptors and ligands, they influence the pathogenesis of various diseases. Recently, a less-explored aspect of neuroimmune communication has emerged: the release of neuropeptides from immune cells and cytokines/chemokines from sensory neurons. This article reviews evidence of this unique neuroimmune interplay and its impact on the development of allergy, inflammation, itch, and pain. We highlight the effects of this neuroimmune signaling on vital processes such as host defense, tissue repair, and inflammation resolution, providing avenues for exploration of the underlying mechanisms and therapeutic potential of this signaling.
Collapse
Affiliation(s)
- Surbhi Gupta
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Alice Viotti
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Tuany Eichwald
- Department of Pharmacology and Physiology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Anais Roger
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Eva Kaufmann
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Rahmeh Othman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Moutih Rafei
- Department of Pharmacology and Physiology, University of Montréal, Montréal, Québec, Canada
| | - Simmie L Foster
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Mass
| | - Sebastien Talbot
- Department of Pharmacology and Physiology, Karolinska Institutet, Solna, Sweden; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
23
|
Wang S, Ko CC, Chung MK. Nociceptor mechanisms underlying pain and bone remodeling via orthodontic forces: toward no pain, big gain. FRONTIERS IN PAIN RESEARCH 2024; 5:1365194. [PMID: 38455874 PMCID: PMC10917994 DOI: 10.3389/fpain.2024.1365194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Orthodontic forces are strongly associated with pain, the primary complaint among patients wearing orthodontic braces. Compared to other side effects of orthodontic treatment, orthodontic pain is often overlooked, with limited clinical management. Orthodontic forces lead to inflammatory responses in the periodontium, which triggers bone remodeling and eventually induces tooth movement. Mechanical forces and subsequent inflammation in the periodontium activate and sensitize periodontal nociceptors and produce orthodontic pain. Nociceptive afferents expressing transient receptor potential vanilloid subtype 1 (TRPV1) play central roles in transducing nociceptive signals, leading to transcriptional changes in the trigeminal ganglia. Nociceptive molecules, such as TRPV1, transient receptor potential ankyrin subtype 1, acid-sensing ion channel 3, and the P2X3 receptor, are believed to mediate orthodontic pain. Neuropeptides such as calcitonin gene-related peptides and substance P can also regulate orthodontic pain. While periodontal nociceptors transmit nociceptive signals to the brain, they are also known to modulate alveolar bone remodeling in periodontitis. Therefore, periodontal nociceptors and nociceptive molecules may contribute to the modulation of orthodontic tooth movement, which currently remains undetermined. Future studies are needed to better understand the fundamental mechanisms underlying neuroskeletal interactions in orthodontics to improve orthodontic treatment by developing novel methods to reduce pain and accelerate orthodontic tooth movement-thereby achieving "big gains with no pain" in clinical orthodontics.
Collapse
Affiliation(s)
- Sheng Wang
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Ching-Chang Ko
- Division of Orthodontics, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Baltimore, MD, United States
- Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, United States
| |
Collapse
|
24
|
Abstract
Interoception, the ability to precisely and timely sense internal body signals, is critical for life. The interoceptive system monitors a large variety of mechanical, chemical, hormonal, and pathological cues using specialized organ cells, organ innervating neurons, and brain sensory neurons. It is important for maintaining body homeostasis, providing motivational drives, and regulating autonomic, cognitive, and behavioral functions. However, compared to external sensory systems, our knowledge about how diverse body signals are coded at a system level is quite limited. In this review, we focus on the unique features of interoceptive signals and the organization of the interoceptive system, with the goal of better understanding the coding logic of interoception.
Collapse
Affiliation(s)
- Ruiqi L Wang
- Department of Neuroscience and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - Rui B Chang
- Department of Neuroscience and Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| |
Collapse
|
25
|
He H, Zhou J, Xu X, Zhou P, Zhong H, Liu M. Piezo channels in the intestinal tract. Front Physiol 2024; 15:1356317. [PMID: 38379701 PMCID: PMC10877011 DOI: 10.3389/fphys.2024.1356317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/24/2024] [Indexed: 02/22/2024] Open
Abstract
The intestine is the largest mechanosensitive organ in the human body whose epithelial cells, smooth muscle cells, neurons and enteroendocrine cells must sense and respond to various mechanical stimuli such as motility, distension, stretch and shear to regulate physiological processes including digestion, absorption, secretion, motility and immunity. Piezo channels are a newly discovered class of mechanosensitive ion channels consisting of two subtypes, Piezo1 and Piezo2. Piezo channels are widely expressed in the intestine and are involved in physiological and pathological processes. The present review summarizes the current research progress on the expression, function and regulation of Piezo channels in the intestine, with the aim of providing a reference for the future development of therapeutic strategies targeting Piezo channels.
Collapse
Affiliation(s)
- Haolong He
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jingying Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xuan Xu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Pinxi Zhou
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Huan Zhong
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Acupuncture and Moxibustion Bioinformatics, Education Department of Hunan Province, Changsha, Hunan, China
| | - Mi Liu
- School of Acupuncture-Moxibustion, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Key Laboratory of Acupuncture and Moxibustion Bioinformatics, Education Department of Hunan Province, Changsha, Hunan, China
| |
Collapse
|
26
|
Zhang X, Wang F, Su Y. TRPV: An emerging target in glaucoma and optic nerve damage. Exp Eye Res 2024; 239:109784. [PMID: 38199261 DOI: 10.1016/j.exer.2024.109784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/30/2023] [Accepted: 01/06/2024] [Indexed: 01/12/2024]
Abstract
Transient receptor potential vanilloid (TRPV) channels are members of the TRP channel superfamily, which are ion channels that sense mechanical and osmotic stimuli and participate in Ca2+ signalling across the cell membrane. TRPV channels play important roles in maintaining the normal functions of an organism, and defects or abnormalities in TRPV channel function cause a range of diseases, including cardiovascular, neurological and urological disorders. Glaucoma is a group of chronic progressive optic nerve diseases with pathological changes that can occur in the tissues of the anterior and posterior segments of the eye, including the ciliary body, trabecular meshwork, Schlemm's canal, and retina. TRPV channels are expressed in these tissues and play various roles in glaucoma. In this article, we review various aspects of the pathogenesis of glaucoma, the structure and function of TRPV channels, the relationship between TRPV channels and systemic diseases, and the relationship between TRPV channels and ocular diseases, especially glaucoma, and we suggest future research directions. This information will help to further our understanding of TRPV channels and provide new ideas and targets for the treatment of glaucoma and optic nerve damage.
Collapse
Affiliation(s)
- Xiaotong Zhang
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Feng Wang
- Department of Ophthalmology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Ying Su
- Eye Hospital, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
27
|
Li YC, Zhang FC, Xu TW, Weng RX, Zhang HH, Chen QQ, Hu S, Gao R, Li R, Xu GY. Advances in the pathological mechanisms and clinical treatments of chronic visceral pain. Mol Pain 2024; 20:17448069241305942. [PMID: 39673493 PMCID: PMC11645724 DOI: 10.1177/17448069241305942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 12/16/2024] Open
Abstract
Chronic visceral pain stems from internal organs and is frequently associated with functional gastrointestinal disorders, like irritable bowel syndrome (IBS). Since the underlying mechanisms of visceral pain remain largely unclear, clinical management is often limited and ineffective. Comprehensive research into the pathogenesis of visceral pain, along with the development of personalized therapeutic strategies, is crucial for advancing treatment options. Studies suggest that imbalances in purinergic receptors and neural circuit function are closely linked to the onset of visceral pain. In this review, we will explore the etiology and pathological mechanisms underlying visceral pain, with a focus on ion channels, epigenetic factors, and neural circuits, using functional gastrointestinal disorders as case studies. Finally, we will summarize and evaluate emerging treatments and potential initiatives aimed at managing visceral pain.
Collapse
Affiliation(s)
- Yong-Chang Li
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fu-Chao Zhang
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Timothy W Xu
- Department of Earth Sciences, University College London, London, UK
| | - Rui-Xia Weng
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong-Hong Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian-Qian Chen
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Shufen Hu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Rong Gao
- Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Guang-Yin Xu
- Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
28
|
Bouchatta O, Brodzki M, Manouze H, Carballo GB, Kindström E, de-Faria FM, Yu H, Kao AR, Thorell O, Liljencrantz J, Ng KKW, Frangos E, Ragnemalm B, Saade D, Bharucha-Goebel D, Szczot I, Moore W, Terejko K, Cole J, Bonnemann C, Luo W, Mahns DA, Larsson M, Gerling GJ, Marshall AG, Chesler AT, Olausson H, Nagi SS, Szczot M. PIEZO2-dependent rapid pain system in humans and mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569650. [PMID: 38168273 PMCID: PMC10760115 DOI: 10.1101/2023.12.01.569650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The PIEZO2 ion channel is critical for transducing light touch into neural signals but is not considered necessary for transducing acute pain in humans. Here, we discovered an exception - a form of mechanical pain evoked by hair pulling. Based on observations in a rare group of individuals with PIEZO2 deficiency syndrome, we demonstrated that hair-pull pain is dependent on PIEZO2 transduction. Studies in control participants showed that hair-pull pain triggered a distinct nocifensive response, including a nociceptive reflex. Observations in rare Aβ deafferented individuals and nerve conduction block studies in control participants revealed that hair-pull pain perception is dependent on Aβ input. Single-unit axonal recordings revealed that a class of cooling-responsive myelinated nociceptors in human skin is selectively tuned to painful hair-pull stimuli. Further, we pharmacologically mapped these nociceptors to a specific transcriptomic class. Finally, using functional imaging in mice, we demonstrated that in a homologous nociceptor, Piezo2 is necessary for high-sensitivity, robust activation by hair-pull stimuli. Together, we have demonstrated that hair-pulling evokes a distinct type of pain with conserved behavioral, neural, and molecular features across humans and mice.
Collapse
Affiliation(s)
- Otmane Bouchatta
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- These authors contributed equally
| | - Marek Brodzki
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- These authors contributed equally
| | - Houria Manouze
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Gabriela B. Carballo
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Emma Kindström
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Felipe M. de-Faria
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Huasheng Yu
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Anika R. Kao
- School of Engineering and Applied Science, University of Virginia, Charlottesville, USA
| | - Oumie Thorell
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- School of Medicine, Western Sydney University, Sydney, Australia
| | - Jaquette Liljencrantz
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, USA
- Department of Anesthesiology and Intensive Care, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Kevin K. W. Ng
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Eleni Frangos
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, USA
| | - Bengt Ragnemalm
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Dimah Saade
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, USA
| | - Diana Bharucha-Goebel
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, USA
| | - Ilona Szczot
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Warren Moore
- Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Katarzyna Terejko
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- Biology of Astrocytes Research Group, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wroclaw, Poland
| | - Jonathan Cole
- University Hospitals, Dorset, and University of Bournemouth, UK
| | - Carsten Bonnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, USA
| | - Wenquin Luo
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - David A. Mahns
- School of Medicine, Western Sydney University, Sydney, Australia
| | - Max Larsson
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Gregory J. Gerling
- School of Engineering and Applied Science, University of Virginia, Charlottesville, USA
| | - Andrew G. Marshall
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- Institute of Life Course and Medical Sciences, University of Liverpool, UK
| | - Alexander T. Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, USA
| | - Håkan Olausson
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
| | - Saad S. Nagi
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- School of Medicine, Western Sydney University, Sydney, Australia
- Senior author
| | - Marcin Szczot
- Center for Social and Affective Neuroscience, Linköping University, Linköping, Sweden
- Senior author
| |
Collapse
|
29
|
Abstract
All cells in the body are exposed to physical force in the form of tension, compression, gravity, shear stress, or pressure. Cells convert these mechanical cues into intracellular biochemical signals; this process is an inherent property of all cells and is essential for numerous cellular functions. A cell's ability to respond to force largely depends on the array of mechanical ion channels expressed on the cell surface. Altered mechanosensing impairs conscious senses, such as touch and hearing, and unconscious senses, like blood pressure regulation and gastrointestinal (GI) activity. The GI tract's ability to sense pressure changes and mechanical force is essential for regulating motility, but it also underlies pain originating in the GI tract. Recent identification of the mechanically activated ion channels Piezo1 and Piezo2 in the gut and the effects of abnormal ion channel regulation on cellular function indicate that these channels may play a pathogenic role in disease. Here, we discuss our current understanding of mechanically activated Piezo channels in the pathogenesis of pancreatic and GI diseases, including pancreatitis, diabetes mellitus, irritable bowel syndrome, GI tumors, and inflammatory bowel disease. We also describe how Piezo channels could be important targets for treating GI diseases.
Collapse
|
30
|
He J, Xie X, Xiao Z, Qian W, Zhang L, Hou X. Piezo1 in Digestive System Function and Dysfunction. Int J Mol Sci 2023; 24:12953. [PMID: 37629134 PMCID: PMC10454946 DOI: 10.3390/ijms241612953] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Piezo1, a non-selective cation channel directly activated by mechanical forces, is widely expressed in the digestive system and participates in biological functions physiologically and pathologically. In this review, we summarized the latest insights on Piezo1’s cellular effect across the entire digestive system, and discussed the role of Piezo1 in various aspects including ingestion and digestion, material metabolism, enteric nervous system, intestinal barrier, and inflammatory response within digestive system. The goal of this comprehensive review is to provide a solid foundation for future research about Piezo1 in digestive system physiologically and pathologically.
Collapse
Affiliation(s)
| | | | | | | | - Lei Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.H.); (X.X.); (Z.X.); (W.Q.)
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (J.H.); (X.X.); (Z.X.); (W.Q.)
| |
Collapse
|
31
|
Obeidat AM, Wood MJ, Adamczyk NS, Ishihara S, Li J, Wang L, Ren D, Bennett DA, Miller RJ, Malfait AM, Miller RE. Piezo2 expressing nociceptors mediate mechanical sensitization in experimental osteoarthritis. Nat Commun 2023; 14:2479. [PMID: 37120427 PMCID: PMC10148822 DOI: 10.1038/s41467-023-38241-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 04/17/2023] [Indexed: 05/01/2023] Open
Abstract
Non-opioid targets are needed for addressing osteoarthritis pain, which is mechanical in nature and associated with daily activities such as walking and climbing stairs. Piezo2 has been implicated in the development of mechanical pain, but the mechanisms by which this occurs remain poorly understood, including the role of nociceptors. Here we show that nociceptor-specific Piezo2 conditional knock-out mice were protected from mechanical sensitization associated with inflammatory joint pain in female mice, joint pain associated with osteoarthritis in male mice, as well as both knee swelling and joint pain associated with repeated intra-articular injection of nerve growth factor in male mice. Single cell RNA sequencing of mouse lumbar dorsal root ganglia and in situ hybridization of mouse and human lumbar dorsal root ganglia revealed that a subset of nociceptors co-express Piezo2 and Ntrk1 (the gene that encodes the nerve growth factor receptor TrkA). These results suggest that nerve growth factor-mediated sensitization of joint nociceptors, which is critical for osteoarthritic pain, is also dependent on Piezo2, and targeting Piezo2 may represent a therapeutic option for osteoarthritis pain control.
Collapse
Affiliation(s)
- Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Matthew J Wood
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Natalie S Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Dongjun Ren
- Department of Pharmacology, Northwestern University, Chicago, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center and Department of Neurological Sciences, Rush University Medical Center, Chicago, USA
| | - Richard J Miller
- Department of Pharmacology, Northwestern University, Chicago, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, USA.
| |
Collapse
|
32
|
Madar J, Tiwari N, Smith C, Sharma D, Shen S, Elmahdi A, Qiao LY. Piezo2 regulates colonic mechanical sensitivity in a sex specific manner in mice. Nat Commun 2023; 14:2158. [PMID: 37061508 PMCID: PMC10105732 DOI: 10.1038/s41467-023-37683-7] [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: 03/15/2022] [Accepted: 03/27/2023] [Indexed: 04/17/2023] Open
Abstract
The mechanosensitive ion channel Piezo2 in mucosa and primary afferents transduces colonic mechanical sensation. Here we show that chemogenetic activation or nociceptor-targeted deletion of Piezo2 is sufficient to regulate colonic mechanical sensitivity in a sex dependent manner. Clozapine N-oxide-induced activation of Piezo2;hM3Dq-expressing sensory neurons evokes colonic hypersensitivity in male mice, and causes dyspnea in female mice likely due to effects on lung sensory neurons. Activation of Piezo2-expressing colonic afferent neurons also induces colonic hypersensitivity in male but not female mice. Piezo2 levels in nociceptive neurons are higher in female than in male mice. We also show that Piezo2 conditional deletion from nociceptive neurons increases body weight growth, slows colonic transits, and reduces colonic mechanosensing in female but not male mice. Piezo2 deletion blocks colonic hypersensitivity in male but not female mice. These results suggest that Piezo2 in nociceptive neurons mediates innocuous colonic mechanosensing in female mice and painful sensation in male mice, suggesting a sexual dimorphism of Piezo2 function in the colonic sensory system.
Collapse
Affiliation(s)
- Jonathan Madar
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Namrata Tiwari
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Cristina Smith
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Divya Sharma
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Shanwei Shen
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Alsiddig Elmahdi
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA
| | - Liya Y Qiao
- Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
33
|
Xu J, McGinnis A, Ji RR. Piezo2 mediates visceral mechanosensation: A new therapeutic target for gut pain? Neuron 2023; 111:450-452. [PMID: 36796326 DOI: 10.1016/j.neuron.2023.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Mechanical distension/stretch in the colon provokes visceral hypersensitivity and pain. In this issue of Neuron, Xie et al. report that mechanosensitive Piezo2 channels, expressed by TRPV1-lineage nociceptors, are involved in visceral mechanical nociception and hypersensitivity.
Collapse
Affiliation(s)
- Jing Xu
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|