1
|
Amedzrovi Agbesi RJ, Chassatte L, Chevalier NR. Smooth Muscle Mechanosensitivity Generates and Maintains Pressure Gradients Across the Intestine. Neurogastroenterol Motil 2024:e14972. [PMID: 39651614 DOI: 10.1111/nmo.14972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/06/2024] [Accepted: 11/17/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND The gut, the ureter, or the Fallopian tube all transport biological fluids by generating trains of propagating smooth muscle constrictions collectively known as peristalsis. These tubes connect body compartments at different pressures. We extend here Poiseuille's experiments on liquid flow in inert tubes to an active, mechanosensitive tube: the intestine. METHODS We use as a miniature myogenic peristaltic pump model, the fetal chicken gut, and measured the flow and contractile wave propagation as a function of the initially applied pressures and pressure gradients. We dissect the molecular pathways of smooth muscle mechanosensitivity by measuring the force generated by gut rings in different pharmacological conditions. RESULTS We demonstrate that smooth muscle contractions in response to stretch or pressure is mediated by L-type Ca2+ channels and IP3 receptors. We show that this positive-feedback mechanosensitive behavior can spontaneously generate pressure gradients across gut segments initially subject to equal pressure; this same mechanism tends to stabilize initially applied pressure gradients; it can act jointly or compete with the pressure gradient induced by directional peristaltic waves. We demonstrate that high pressure differentials can reverse the physiological propagation direction of contractile waves imparted by interstitial cell of Cajal pacemaker activity. We find that flow rate increases with tube length, but that the maximum pressure differential generated depends solely on smooth muscle contractile force and on the initial resting pressure applied inside the organ. CONCLUSIONS We provide fundamental mechanical and hydrodynamic insight into the myogenic mechanisms of transport in the gastrointestinal tract. We scale up our results to other human peristaltic organs and discuss their implications for pathophysiology of intestinal obstruction, vesicoureteral reflux and endometriosis.
Collapse
Affiliation(s)
| | - Lucas Chassatte
- Laboratoire Matière et Systèmes Complexes UMR 7057, Université Paris Cité/CNRS, Paris, France
| | - Nicolas R Chevalier
- Laboratoire Matière et Systèmes Complexes UMR 7057, Université Paris Cité/CNRS, Paris, France
| |
Collapse
|
2
|
Arndorfer D, Pandolfino JE, Kahrilas PJ, Carlson DA. Age Affects Esophageal Secondary Peristalsis More Than Primary as Assessed by FLIP Panometry and High-resolution Manometry. Clin Gastroenterol Hepatol 2024; 22:2532-2534.e1. [PMID: 38866171 PMCID: PMC11588548 DOI: 10.1016/j.cgh.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
Age-related changes in esophageal function involve increased rates of dysmotility with advancing age (sometimes described as presbyesophagus).1-4 Specifically, advanced age has been associated with increased lower esophageal sphincter (LES) pressure and decreased peristaltic vigor.1-4 However, the mechanisms underlying these changes, including the relative relationship of primary peristalsis and secondary peristalsis over the lifespan, remain incompletely understood. Current approaches assess primary peristalsis using high-resolution manometry (HRM), with esophageal motility disorders defined using the Chicago Classification version 4.0 (CCv4.0), and secondary peristalsis using functional lumen imaging probe (FLIP) panometry.5,6 Although HRM and FLIP panometry motility diagnoses are often analogous, they can sometimes differ. For example, abnormal secondary peristalsis can occur when primary peristalsis is normal, which has been associated with reflux and non-obstructive dysphagia.7,8 This study aimed to examine the effect of age on primary peristalsis and secondary peristalsis.
Collapse
Affiliation(s)
- Daniel Arndorfer
- Kenneth C. Griffin Esophageal Center of Northwestern Medicine, Department of Medicine, Division of Gastroenterology and Hepatology; Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - John E Pandolfino
- Kenneth C. Griffin Esophageal Center of Northwestern Medicine, Department of Medicine, Division of Gastroenterology and Hepatology; Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Peter J Kahrilas
- Kenneth C. Griffin Esophageal Center of Northwestern Medicine, Department of Medicine, Division of Gastroenterology and Hepatology; Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Dustin A Carlson
- Kenneth C. Griffin Esophageal Center of Northwestern Medicine, Department of Medicine, Division of Gastroenterology and Hepatology; Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
3
|
Jiang Q, Li Z, Dang D, Wei J, Wu H. Role of mechanosensitive channel Piezo1 protein in intestinal inflammation regulation: A potential target. FASEB J 2024; 38:e70122. [PMID: 39425504 PMCID: PMC11580726 DOI: 10.1096/fj.202401323r] [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: 06/11/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
The intestine is a hollow tract that primarily transports and digests food. It often encounters mechanical forces and exotic threats, resulting in increased intestinal inflammation attributed to the consistent threat of foreign pathogens. Piezo1, a mechanosensitive ion channel, is distributed broadly and abundantly in the intestinal tissue. It transduces mechanical signals into electrochemical signals and participates in many critical life activities, such as proliferation, differentiation, cell apoptosis, immune cell activation, and migration. Its effect on inflammation has been discussed in detail in systems, such as musculoskeletal (osteoarthritis) and cardiac (myocarditis), but the effects on intestinal inflammation remain unelucidated. Piezo1 regulates mucosal layer and epithelial barrier homeostasis during the complex intestinal handling of foreign antigens and tissue trauma. It initiates and spreads immune responses and causes distant effects of inflammation in the vascular and lymphatic systems, but reports of the effects of Piezo1 in intestinal inflammation are scarce. Therefore, this study aimed to discuss the role of Piezo1 in intestinal inflammation and explore novel therapeutic targets.
Collapse
Affiliation(s)
- Qinlei Jiang
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Zhenyu Li
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Dan Dang
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Jiaqi Wei
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| | - Hui Wu
- Department of Neonatology, Children's Medical CenterThe First Hospital of Jilin UniversityChangchunJilinPeople's Republic of China
| |
Collapse
|
4
|
Liu S, Xi H, Xue X, Sun X, Huang H, Fu D, Mi Y, He Y, Yang P, Tang Y, Zheng P. Clostridium butyricum regulates intestinal barrier function via trek1 to improve behavioral abnormalities in mice with autism spectrum disorder. Cell Biosci 2024; 14:95. [PMID: 39034406 PMCID: PMC11265103 DOI: 10.1186/s13578-024-01278-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder that has been found to be associated with dysregulation of gastrointestinal functions and gut microbial homeostasis (the so-called "gut-brain axis"). ASD is often accompanied by poor performances in social interaction and repetitive behaviors. Studies on the gut-brain axis provide novel insights and candidate targets for ASD therapeutics and diagnosis. Based on the ASD mice model, this work aims to reveal the mechanisms behind the interaction of intestinal barrier function and probiotics in ASD mouse models. RESULTS We found an altered intestinal barrier in both BTBR T+ Itpr3tf/J (BTBR) and valproic acid (VPA) mice, including increased intestinal permeability, decreased expression of intestinal tight junction proteins (claudin1, claudin3, and occludin), and increased levels of IL-6, TNF-α, and IFN-γ. Based on intestinal microbial alternation, C. butyricum can drive reduced expression of histone deacetylases 1 (HDAC1) and enhanced intestinal barrier function, significantly promoting behavioral abnormalities of ASD in BTBR mice. In parallel, we confirmed that C. butyricum was involved in the regulation of intestinal function by the Trek1 channel, indicating that it is a target of C. butyricum/butyric acid to improve intestinal barrier function in ASD mice. CONCLUSIONS Our finding provides solid evidence for the gut microbiota involved in ASD through the brain-gut axis. In addition, the probiotics C. butyricum hold promise to improve gut health and ameliorate behavioral abnormalities associated with ASD.
Collapse
Affiliation(s)
- Simeng Liu
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Xiangyu Medical CO., LTD, Anyang, 456300, Henan, China.
| | - Huayuan Xi
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xia Xue
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiangdong Sun
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Huang Huang
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Dongjun Fu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Mi
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yongzheng He
- Xiangyu Medical CO., LTD, Anyang, 456300, Henan, China
| | - Pingchang Yang
- Brain Body Institute, McMaster University, Hamilton, ON, Canada
| | - Youcai Tang
- Department of Pediatrics, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Pengyuan Zheng
- Marshall B. J. Medical Research Center, Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Gastroenterology, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
5
|
Yao YB, Xiao CF, Wu JW, Meng LY, Liu W, Lu JG, Wang C. Yiqi Kaimi prescription regulates protein phosphorylation to promote intestinal motility in slow transit constipation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118118. [PMID: 38614261 DOI: 10.1016/j.jep.2024.118118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/23/2024] [Accepted: 03/26/2024] [Indexed: 04/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The clinical efficacy of the Yiqi Kaimi prescription has been confirmed in slow transit constipation. However, the effects and biological mechanism of Yiqi Kaimi prescription are still unclear. AIMS OF THE STUDY To identify the effects of Yiqi Kaimi prescription on intestinal motility; To reveal the potential key targets and pathways of Yiqi Kaimi prescription for the treatment of slow transit constipation. MATERIALS AND METHODS The effects of Yiqi Kaimi prescription on slow transit constipation were investigated in a mouse model. The terminal ink propulsion experiment and fecal indocyanine green imaging was used to measure the intestinal transit time. Protein phosphorylation changes in colon tissues treated with Yiqi Kaimi prescription were detected using a Phospho Explorer antibody microarray. Bioinformatic analyses were performed using the Database for Annotation Visualization and Integrated Discovery (DAVID) and the Search Tool for the Retrieval of Interacting Genes (STRING). Western blot analysis and immunohistochemistry confirmed the observed changes in phosphorylation. RESULT s: Yiqi Kaimi prescription significantly increased the intestinal transit rate (P < 0.05 vs. model) and reduced the time to first discharge of feces containing fecal indocyanine green imaging in mice (P < 0.05 vs. model). The administration of Yiqi Kaimi prescription induced phosphorylation changes in 41 proteins, with 9 upregulated proteins and 32 downregulated proteins. Functional classification of the phosphorylated proteins with DAVID revealed that the critical biological processes included tyrosine protein kinases, positive regulation of calcium-mediated signaling and response to muscle stretch. The phosphorylation of the spleen tyrosine kinase (SYK) at Tyr348 increased 2.19-fold, which was the most significant change. The phosphorylation level of the transcription factor p65 (RELA) at Thr505 was decreased 0.57-fold. SYK was a hub protein in the protein-protein interaction network and SYK and RELA formed the core of the secondary subnetwork. The key protein phosphorylation after treatment with Yiqi Kaimi prescription were verified by Western blot analysis and immunohistochemistry. CONCLUSION Yiqi Kaimi prescription significantly enhanced intestinal motility. This effect was attributed to alterations in the phosphorylation levels of various target proteins. The observed changes in protein phosphorylation, including SYK and RELA, may serve as crucial factors in the treatment of slow transit constipation.
Collapse
Affiliation(s)
- Yi-Bo Yao
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| | - Chang-Fang Xiao
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Jing-Wen Wu
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Ling-Yun Meng
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Wei Liu
- Department of Pharmacy, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jin-Gen Lu
- Institute of Chinese Traditional Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Chen Wang
- Department of Anorectal Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| |
Collapse
|
6
|
Ouyang M, Zhang Q, Zhu Y, Luo M, Bu B, Deng L. α-Catenin and Piezo1 Mediate Cell Mechanical Communication via Cell Adhesions. BIOLOGY 2024; 13:357. [PMID: 38785839 PMCID: PMC11118126 DOI: 10.3390/biology13050357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Cell-to-cell distant mechanical communication has been demonstrated using in vitro and in vivo models. However, the molecular mechanisms underlying long-range cell mechanoresponsive interactions remain to be fully elucidated. This study further examined the roles of α-Catenin and Piezo1 in traction force-induced rapid branch assembly of airway smooth muscle (ASM) cells on a Matrigel hydrogel containing type I collagen. Our findings demonstrated that siRNA-mediated downregulation of α-Catenin or Piezo1 expression or chemical inhibition of Piezo1 activity significantly reduced both directional cell movement and branch assembly. Regarding the role of N-cadherin in regulating branch assembly but not directional migration, our results further confirmed that siRNA-mediated downregulation of α-Catenin expression caused a marked reduction in focal adhesion formation, as assessed by focal Paxillin and Integrin α5 localization. These observations imply that mechanosensitive α-Catenin is involved in both cell-cell and cell-matrix adhesions. Additionally, Piezo1 partially localized in focal adhesions, which was inhibited by siRNA-mediated downregulation of α-Catenin expression. This result provides insights into the Piezo1-mediated mechanosensing of traction force on a hydrogel. Collectively, our findings highlight the significance of α-Catenin in the regulation of cell-matrix interactions and provide a possible interpretation of Piezo1-mediated mechanosensing activity at focal adhesions during cell-cell mechanical communication.
Collapse
Affiliation(s)
- Mingxing Ouyang
- Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China (M.L.); (B.B.)
| | - Qingyu Zhang
- Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China (M.L.); (B.B.)
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yiming Zhu
- Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China (M.L.); (B.B.)
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Mingzhi Luo
- Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China (M.L.); (B.B.)
| | - Bing Bu
- Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China (M.L.); (B.B.)
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China (M.L.); (B.B.)
| |
Collapse
|
7
|
Goyal RK, Rattan S. Role of mechanoregulation in mast cell-mediated immune inflammation of the smooth muscle in the pathophysiology of esophageal motility disorders. Am J Physiol Gastrointest Liver Physiol 2024; 326:G398-G410. [PMID: 38290993 PMCID: PMC11213482 DOI: 10.1152/ajpgi.00258.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/01/2024]
Abstract
Major esophageal disorders involve obstructive transport of bolus to the stomach, causing symptoms of dysphagia and impaired clearing of the refluxed gastric contents. These may occur due to mechanical constriction of the esophageal lumen or loss of relaxation associated with deglutitive inhibition, as in achalasia-like disorders. Recently, immune inflammation has been identified as an important cause of esophageal strictures and the loss of inhibitory neurotransmission. These disorders are also associated with smooth muscle hypertrophy and hypercontractility, whose cause is unknown. This review investigated immune inflammation in the causation of smooth muscle changes in obstructive esophageal bolus transport. Findings suggest that smooth muscle hypertrophy occurs above the obstruction and is due to mechanical stress on the smooth muscles. The mechanostressed smooth muscles release cytokines and other molecules that may recruit and microlocalize mast cells to smooth muscle bundles, so that their products may have a close bidirectional effect on each other. Acting in a paracrine fashion, the inflammatory cytokines induce genetic and epigenetic changes in the smooth muscles, leading to smooth muscle hypercontractility, hypertrophy, and impaired relaxation. These changes may worsen difficulty in the esophageal transport. Immune processes differ in the first phase of obstructive bolus transport, and the second phase of muscle hypertrophy and hypercontractility. Moreover, changes in the type of mechanical stress may change immune response and effect on smooth muscles. Understanding immune signaling in causes of obstructive bolus transport, type of mechanical stress, and associated smooth muscle changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.NEW & NOTEWORTHY Esophageal disorders such as esophageal stricture or achalasia, and diffuse esophageal spasm are associated with smooth muscle hypertrophy and hypercontractility, above the obstruction, yet the cause of such changes is unknown. This review suggests that smooth muscle obstructive disorders may cause mechanical stress on smooth muscle, which then secretes chemicals that recruit, microlocalize, and activate mast cells to initiate immune inflammation, producing functional and structural changes in smooth muscles. Understanding the immune signaling in these changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.
Collapse
Affiliation(s)
- Raj K Goyal
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Boston Healthcare System, West Roxbury, Massachusetts, United States
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts, United States
| | - Satish Rattan
- Department of Medicine, Division of Gastroenterology and Hepatology, Sidney Kummel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
8
|
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
|
9
|
Tearle JLE, Tang A, Vasanthakumar A, James KR. Role reversals: non-canonical roles for immune and non-immune cells in the gut. Mucosal Immunol 2024; 17:137-146. [PMID: 37967720 DOI: 10.1016/j.mucimm.2023.11.004] [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: 08/17/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023]
Abstract
The intestine is home to an intertwined network of epithelial, immune, and neuronal cells as well as the microbiome, with implications for immunity, systemic metabolism, and behavior. While the complexity of this microenvironment has long since been acknowledged, recent technological advances have propelled our understanding to an unprecedented level. Notably, the microbiota and non-immune or structural cells have emerged as important conductors of intestinal immunity, and by contrast, cells of both the innate and adaptive immune systems have demonstrated non-canonical roles in tissue repair and metabolism. This review highlights recent works in the following two streams: non-immune cells of the intestine performing immunological functions; and traditional immune cells exhibiting non-immune functions in the gut.
Collapse
Affiliation(s)
- Jacqueline L E Tearle
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Biomedical Sciences, University of New South Wales, Australia
| | - Adelynn Tang
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia
| | - Ajithkumar Vasanthakumar
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Australia; Department of Microbiology and Immunology, University of Melbourne, Melbourne, Australia.
| | - Kylie R James
- Garvan Institute of Medical Research, Darlinghurst, Australia; School of Biomedical Sciences, University of New South Wales, Australia.
| |
Collapse
|
10
|
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
|
11
|
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: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
12
|
Maher S, Bayachou M, Fu P, Hijaz A, Liu G. Focal adhesion kinase activation is involved in contractile stimulation-induced detrusor muscle contraction in mice. Eur J Pharmacol 2023; 952:175807. [PMID: 37236435 PMCID: PMC10330804 DOI: 10.1016/j.ejphar.2023.175807] [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: 03/13/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Recent studies suggested smooth muscle contraction may involve mechanisms besides the myosin regulatory light chain (MLC) phosphorylation-induced actomyosin crossbridge cycling. This study aims to determine if focal adhesion kinase (FAK) activation is involved in mouse detrusor muscle contraction. The mouse detrusor muscle strips were preincubated with PF-573228 (2 μM), latrunculin B (1 μM), or the same volume of vehicle (DMSO) for 30 min. The contractile responses to KCl (90 mM), electrical field stimulation (EFS, 2-32 Hz), or carbachol (CCh, 10-7.5-10-4.5 M) were measured. In a separate experiment, the phosphorylated FAK (p-FAK) and MLC (p-MLC) levels were measured in the detrusor strips stimulated with CCh (10 μM) after incubation with PF-573228 or vehicle (DMSO) compared to those with vehicle incubation but without CCh stimulation. KCl-induced contractile responses decreased significantly after incubation with PF-573228 or latrunculin B compared to the corresponding vehicle-treated strips (p < 0.0001). The contractile responses induced by EFS were markedly inhibited by preincubation with PF-573228 at 8, 16, and 32 Hz (p < 0.05) or latrunculin B at 16 and 32 Hz (p < 0.01). Following the application of PF-573228 or Latrunculin B, CCh-induced dose-response contractions were lower than the corresponding vehicle group (p = 0.0021 and 0.0003, respectively). Western blot examination showed that CCh stimulation enhanced the expression of p-FAK and p-MLC, while preincubation with PF-573228 prevented the increase of p-FAK but not p-MLC. In conclusion, FAK activation involves tension development induced by contractile stimulation in the mouse detrusor muscle. This effect is likely caused by promoting actin polymerization rather than elevating MLC phosphorylation.
Collapse
Affiliation(s)
- Shaimaa Maher
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA; Department of Chemistry, Cleveland State University, Cleveland, OH, USA
| | - Mekki Bayachou
- Department of Chemistry, Cleveland State University, Cleveland, OH, USA; Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Adonis Hijaz
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Guiming Liu
- Department of Surgery, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
13
|
Naing LY, Mathur P, Chhabra G, Stocker A, Abell TL. Gastroparesis and Gastroparesis Syndromes as Neuromuscular Disorders. Semin Neurol 2023; 43:540-552. [PMID: 37562455 DOI: 10.1055/s-0043-1771469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Gastroparesis syndromes (GpS) are a spectrum of disorders presenting with characteristic symptoms increasingly recognized as being gastrointestinal (GI) neuromuscular disorders (NMDs). This review focuses on GpS as a manifestation of neurologic disorders of GI NMD. GpS can be associated with systemic abnormalities, including inflammatory, metabolic, and serologic disorders, as well as autoimmune antibodies via nerve and muscle targets in the GI tract, which can be treated with immunotherapy, such as intravenous immunoglobulin. GpS are associated with autonomic (ANS) and enteric (ENS) dysfunction. Disorders of ANS may interact with the ENS and are the subject of continued investigation. ENS disorders have been recognized for a century but have only recently begun to be fully quantified. Anatomic structural changes in the GI tract are increasingly recognized in GpS. Detailed descriptions of anatomic changes in GpS, and their correlation with physiologic findings, have opened a new era of investigation. The management of GpS, when viewed as GI NMD, has shifted the paradigms of both diagnosis and treatment. This article concludes with current approaches to GpS directed at underlying neuromuscular pathology.
Collapse
Affiliation(s)
- Le Yu Naing
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Prateek Mathur
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Garvit Chhabra
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Abigail Stocker
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Thomas L Abell
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
14
|
Khoshavi Najafabadi F, Sadraei H, Mehranfard N, Ghasemi M. Motor Dysfunction of Gastric Antral Smooth Muscle in Diabetic Rats: Contribution of ATP-Dependent Potassium Channels. Adv Biomed Res 2023; 12:199. [PMID: 37694236 PMCID: PMC10492619 DOI: 10.4103/abr.abr_44_23] [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: 02/06/2023] [Revised: 05/14/2023] [Accepted: 05/20/2023] [Indexed: 09/12/2023] Open
Abstract
Background The goal of the current research was to further elucidate the role of adenosine triphosphate (ATP)-sensitive potassium (KATP) channels in the motility and contractility force of gastric smooth muscle of diabetic rats. Materials and Methods Male Wistar rats (190-230 g) were grouped into control and streptozotocin (STZ)-induced diabetes (55 mg/kg) rats. Thirty days later, gastric muscle contractility was measured using a myograph and a force transducer of antral segments immersed in a tissue bath. Gastric emptying response was measured through feeding of standard pellet. Furthermore, the expression of KATP channel subunits in antral smooth muscle was determined by western blot technique. Results The amplitude of KCl-evoked twitch contractions of diabetic antral strips was about 25% more than control (P < 0.05). Application of minoxidil, a KATP channel opener, dose dependently decreased the force of twitch contractions in both normal and diabetic antral strips. Application of 10 μM glibenclamide, a KATP channel blocker, did not antagonize the minoxidil-induced relaxation of antral strips. Diabetic gastric emptying was faster than normal, although not significant. Despite the relaxant effect of minoxidil on gastric emptying rate in normal rats (P < 0.05), this effect was not observed in diabetic rats. Also, glibenclamide increased gastric emptying and antagonized minoxidil-induced relaxation in normal rats (P < 0.05). Furthermore, the expression of KATP Kir6.1 and SUR2B subunits was substantially reduced in antral smooth muscle in diabetic condition (P < 0.01). Conclusion These results propose that KATP channels may contribute to the development of gastric motility disorders in diabetes.
Collapse
Affiliation(s)
- Fatameh Khoshavi Najafabadi
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Sadraei
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Mehranfard
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
15
|
Remme CA. SCN5A channelopathy: arrhythmia, cardiomyopathy, epilepsy and beyond. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220164. [PMID: 37122208 PMCID: PMC10150216 DOI: 10.1098/rstb.2022.0164] [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] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/31/2022] [Indexed: 05/02/2023] Open
Abstract
Influx of sodium ions through voltage-gated sodium channels in cardiomyocytes is essential for proper electrical conduction within the heart. Both acquired conditions associated with sodium channel dysfunction (myocardial ischaemia, heart failure) as well as inherited disorders secondary to mutations in the gene SCN5A encoding for the cardiac sodium channel Nav1.5 are associated with life-threatening arrhythmias. Research in the last decade has uncovered the complex nature of Nav1.5 distribution, function, in particular within distinct subcellular subdomains of cardiomyocytes. Nav1.5-based channels furthermore display previously unrecognized non-electrogenic actions and may impact on cardiac structural integrity, leading to cardiomyopathy. Moreover, SCN5A and Nav1.5 are expressed in cell types other than cardiomyocytes as well as various extracardiac tissues, where their functional role in, e.g. epilepsy, gastrointestinal motility, cancer and the innate immune response is increasingly investigated and recognized. This review provides an overview of these novel insights and how they deepen our mechanistic knowledge on SCN5A channelopathies and Nav1.5 (dys)function. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Carol Ann Remme
- Department of Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC location AMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Caulk AW, Chatterjee M, Barr SJ, Contini EM. Mechanobiological considerations in colorectal stapling: Implications for technology development. Surg Open Sci 2023; 13:54-65. [PMID: 37159635 PMCID: PMC10163679 DOI: 10.1016/j.sopen.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/07/2023] [Accepted: 04/08/2023] [Indexed: 05/11/2023] Open
Abstract
Technological advancements in minimally invasive surgery have led to significant improvements in patient outcomes. One such technology is surgical stapling, which has evolved into a key component of many operating rooms by facilitating ease and efficacy in resection and repair of diseased or otherwise compromised tissue. Despite such advancements, adverse post-operative outcomes such as anastomotic leak remain a persistent problem in surgical stapling and its correlates (i.e., hand-sewing), most notably in low colorectal or coloanal procedures. Many factors may drive anastomotic leaks, including tissue perfusion, microbiome composition, and patient factors such as pre-existing disease. Surgical intervention induces complex acute and chronic changes to the mechanical environment of the tissue; however, roles of mechanical forces in post-operative healing remain poorly characterized. It is well known that cells sense and respond to their local mechanical environment and that dysfunction of this "mechanosensing" phenomenon contributes to a myriad of diseases. Mechanosensing has been investigated in wound healing contexts such as dermal incisional and excisional wounds and development of pressure ulcers; however, reports investigating roles of mechanical forces in adverse post-operative gastrointestinal wound healing are lacking. To understand this relationship well, it is critical to understand: 1) the intraoperative material responses of tissue to surgical intervention, and 2) the post-operative mechanobiological response of the tissue to surgically imposed forces. In this review, we summarize the state of the field in each of these contexts while highlighting areas of opportunity for discovery and innovation which can positively impact patient outcomes in minimally invasive surgery.
Collapse
|
17
|
Abstract
A large body of evidence has emerged in the past decade supporting a role for the gut microbiome in the regulation of blood pressure. The field has moved from association to causation in the last 5 years, with studies that have used germ-free animals, antibiotic treatments and direct supplementation with microbial metabolites. The gut microbiome can regulate blood pressure through several mechanisms, including through gut dysbiosis-induced changes in microbiome-associated gene pathways in the host. Microbiota-derived metabolites are either beneficial (for example, short-chain fatty acids and indole-3-lactic acid) or detrimental (for example, trimethylamine N-oxide), and can activate several downstream signalling pathways via G protein-coupled receptors or through direct immune cell activation. Moreover, dysbiosis-associated breakdown of the gut epithelial barrier can elicit systemic inflammation and disrupt intestinal mechanotransduction. These alterations activate mechanisms that are traditionally associated with blood pressure regulation, such as the renin-angiotensin-aldosterone system, the autonomic nervous system, and the immune system. Several methodological and technological challenges remain in gut microbiome research, and the solutions involve minimizing confounding factors, establishing causality and acting globally to improve sample diversity. New clinical trials, precision microbiome medicine and computational methods such as Mendelian randomization have the potential to enable leveraging of the microbiome for translational applications to lower blood pressure.
Collapse
|
18
|
Du P, Joshi V, Beyder A. Tracking Gut Motility in Organ and Cultures. Methods Mol Biol 2023; 2644:449-466. [PMID: 37142940 DOI: 10.1007/978-1-0716-3052-5_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Gastrointestinal (GI) motility is a key component of digestive health, and it is complex, involving a multitude of cell types and mechanisms to drive both rhythmic and arrhythmic activity. Tracking GI motility in organ and tissue cultures across multiple temporal (seconds, minutes, hours, days) scales can provide valuable information regarding dysmotility and to evaluate treatment options. Here, the chapter describes a simple method to monitor GI motility in organotypic cultures, using a single video camera is placed perpendicularly to the surface of the tissue. A cross-correlational analysis is used to track the relative movements of tissues between subsequent frames and subsequent fitting procedures to fit finite element functions to the deformed tissue to calculate the strain fields. Additional motility index measures from the displacement information are used to further quantify the behaviors of the tissues that are maintained in organotypic culture over days. The protocols presented in this chapter can be adapted to study organotypic cultures from other organs.
Collapse
Affiliation(s)
- Peng Du
- Auckland Bioengineering Institute, Department of Engineering Science and Biomedical Engineering, University of Auckland, Auckland, New Zealand.
| | - Vikram Joshi
- Department of Physiology and Biomedical Engineering, Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
19
|
Kola JB, Docsa T, Uray K. Mechanosensing in the Physiology and Pathology of the Gastrointestinal Tract. Int J Mol Sci 2022; 24:ijms24010177. [PMID: 36613619 PMCID: PMC9820522 DOI: 10.3390/ijms24010177] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/10/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Normal gastrointestinal function relies on sensing and transducing mechanical signals into changes in intracellular signaling pathways. Both specialized mechanosensing cells, such as certain enterochromaffin cells and enteric neurons, and non-specialized cells, such as smooth muscle cells, interstitial cells of Cajal, and resident macrophages, participate in physiological and pathological responses to mechanical signals in the gastrointestinal tract. We review the role of mechanosensors in the different cell types of the gastrointestinal tract. Then, we provide several examples of the role of mechanotransduction in normal physiology. These examples highlight the fact that, although these responses to mechanical signals have been known for decades, the mechanosensors involved in these responses to mechanical signals are largely unknown. Finally, we discuss several diseases involving the overstimulation or dysregulation of mechanotransductive pathways. Understanding these pathways and identifying the mechanosensors involved in these diseases may facilitate the identification of new drug targets to effectively treat these diseases.
Collapse
Affiliation(s)
- Job Baffin Kola
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, 4032 Debrecen, Hungary
| | - Tibor Docsa
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, 4032 Debrecen, Hungary
| | - Karen Uray
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Center of Excellence, The Hungarian Academy of Sciences, 4032 Debrecen, Hungary
- Correspondence:
| |
Collapse
|
20
|
Cahusac PM, Senok SS. Effects of potassium channel modulators on the responses of mammalian slowly adapting mechanoreceptors. IBRO Neurosci Rep 2022; 13:344-355. [PMID: 36274789 PMCID: PMC9582710 DOI: 10.1016/j.ibneur.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
Abstract
Introduction slowly adapting mechanoreceptors in the skin provide vital tactile information to animals. The ionic channels that underlie their functioning is the subject of intense research. Previous work suggests that potassium channels may play particular roles in the activation and firing of these mechanoreceptors. Objective We used a range of potassium channel blockers and openers to observe their effects on different phases of mechanoreceptor responses. Methods Extracellular recording of neural activity of slowly adapting mechanoreceptors was carried out in an in vitro preparation of the sinus hair follicles taken from rat whisker pads. A range of potassium (K+) channel modulators were tested on these mechanoreceptor responses. The channel blockers tested were: tetraethylammonium (TEA), barium chloride (BaCl2), dequalinium, 4-aminopyridine (4-AP), paxilline, XE 991, apamin, and charybdotoxin. Results Except for charybdotoxin and apamin, these drugs increased the activity of both types of slowly adapting units, St I and St II. Generally, both spontaneous and evoked (dynamic and static) activities increased. The channel opener NS1619 was also tested. NS1619 clearly decreased evoked activity (both dynamic and static) while leaving spontaneous activity relatively unaffected, with no clear discrimination of effects on the two types of St receptor Conclusion These findings are consistent with the targets of the drugs suggesting that K+ channels play an important role in the maintenance of spontaneous firing and in the production of and persistence of mechanoreceptor activity.
Collapse
Affiliation(s)
- Peter M.B. Cahusac
- College of Medicine, Alfaisal University, Saudi Arabia, and Department of Comparative Medicine, King Faisal Specialist Hospital & Research Centre, PO Box 50927, Riyadh 11533, Kingdom of Saudi Arabia,Correspondence to: Department of Pharmacology & Biostatistics College of Medicine Alfaisal University, PO Box 50927, Riyadh 11533, Kingdom of Saudi Arabia.
| | - Solomon S. Senok
- Ajman University College of Medicine, PO Box 346, Ajman, United Arab Emirates
| |
Collapse
|
21
|
Gut microbiota: a new avenue to reveal pathological mechanisms of constipation. Appl Microbiol Biotechnol 2022; 106:6899-6913. [PMID: 36190540 DOI: 10.1007/s00253-022-12197-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
Constipation is very pervasive all over the world. It is a common multifactorial gastrointestinal disease, and its etiology and pathomechanism are not completely clear. Now, increasing evidence shows that intestinal flora is closely related to constipation. Intestinal flora is the largest microbiota in the human body and has powerful metabolic functions. Intestinal flora can produce a variety of metabolites, such as bile acids, short-chain fatty acids, tryptophan metabolites, and methane, which have important effects on intestinal motility and secretion. The host can also monitor the intestinal flora and regulate gut dysbacteriosis in constipation. To explore the relationship between intestinal flora and host, the combination of multiomics technology has become the powerful and effective method. Furthermore, the homeostasis restoration of intestinal flora also provides a new strategy for the treatment of constipation. This review aims to explore the interaction between intestinal flora and host in constipation, which contributes to disclose the pathogenesis of constipation and the development of novel drugs for the treatment of constipation from the perspective of intestinal flora. KEY POINTS: • This review highlights the regulation of gut microbiota on the intestinal motility and secretion of host. • The current review gives an insight into the role of the host on the recognition and regulation of intestinal ecology under constipation. • The article also introduces some novel methods of current gut microbiota research and gut microbiota-based constipation therapies.
Collapse
|
22
|
Role of Ion Channels in the Chemotransduction and Mechanotransduction in Digestive Function and Feeding Behavior. Int J Mol Sci 2022; 23:ijms23169358. [PMID: 36012643 PMCID: PMC9409042 DOI: 10.3390/ijms23169358] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The gastrointestinal tract constantly communicates with the environment, receiving and processing a wide range of information. The contents of the gastrointestinal tract and the gastrointestinal tract generate mechanical and chemical signals, which are essential for regulating digestive function and feeding behavior. There are many receptors here that sense intestinal contents, including nutrients, microbes, hormones, and small molecule compounds. In signal transduction, ion channels are indispensable as an essential component that can generate intracellular ionic changes or electrical signals. Ion channels generate electrical activity in numerous neurons and, more importantly, alter the action of non-neurons simply and effectively, and also affect satiety, molecular secretion, intestinal secretion, and motility through mechanisms of peripheral sensation, signaling, and altered cellular function. In this review, we focus on the identity of ion channels in chemosensing and mechanosensing in the gastrointestinal tract.
Collapse
|
23
|
Yang H, Hou C, Xiao W, Qiu Y. The role of mechanosensitive ion channels in the gastrointestinal tract. Front Physiol 2022; 13:904203. [PMID: 36060694 PMCID: PMC9437298 DOI: 10.3389/fphys.2022.904203] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanosensation is essential for normal gastrointestinal (GI) function, and abnormalities in mechanosensation are associated with GI disorders. There are several mechanosensitive ion channels in the GI tract, namely transient receptor potential (TRP) channels, Piezo channels, two-pore domain potassium (K2p) channels, voltage-gated ion channels, large-conductance Ca2+-activated K+ (BKCa) channels, and the cystic fibrosis transmembrane conductance regulator (CFTR). These channels are located in many mechanosensitive intestinal cell types, namely enterochromaffin (EC) cells, interstitial cells of Cajal (ICCs), smooth muscle cells (SMCs), and intrinsic and extrinsic enteric neurons. In these cells, mechanosensitive ion channels can alter transmembrane ion currents in response to mechanical forces, through a process known as mechanoelectrical coupling. Furthermore, mechanosensitive ion channels are often associated with a variety of GI tract disorders, including irritable bowel syndrome (IBS) and GI tumors. Mechanosensitive ion channels could therefore provide a new perspective for the treatment of GI diseases. This review aims to highlight recent research advances regarding the function of mechanosensitive ion channels in the GI tract. Moreover, it outlines the potential role of mechanosensitive ion channels in related diseases, while describing the current understanding of interactions between the GI tract and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Haoyu Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Chaofeng Hou
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
- *Correspondence: Yuan Qiu,
| |
Collapse
|
24
|
Abstract
The vertebrate intestine experiences a range of intrinsically generated and external forces during both development and adult homeostasis. It is increasingly understood how the coordination of these forces shapes the intestine through organ-scale folding and epithelial organization into crypt-villus compartments. Moreover, accumulating evidence shows that several cell types in the adult intestine can sense and respond to forces to regulate key cellular processes underlying adult intestinal functions and self-renewal. In this way, transduction of forces may direct both intestinal homeostasis as well as adaptation to external stimuli, such as food ingestion or injury. In this review, we will discuss recent insights from complementary model systems into the force-dependent mechanisms that establish and maintain the unique architecture of the intestine, as well as its homeostatic regulation and function throughout adult life.
Collapse
|
25
|
Abulmeaty MMA, Aldisi D, Aljuraiban GS, Almajwal A, El Shorbagy E, Almuhtadi Y, Albaran B, Aldossari Z, Alsager T, Razak S, Berika M, Al Zaben M. Association of Gastric Myoelectrical Activity With Ghrelin, Gastrin, and Irisin in Adults With Metabolically Healthy and Unhealthy Obesity. Front Physiol 2022; 13:815026. [PMID: 35547577 PMCID: PMC9081643 DOI: 10.3389/fphys.2022.815026] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/22/2022] [Indexed: 12/03/2022] Open
Abstract
Background and Objective: Functional disturbances of gastric myoelectrical activity (GMA) might exist in obesity. However, studies on its association with the gastric hormones in obesity phenotypes are lacking. The objective was to study the association of GMA with the serum levels of key gastric hormones in different obesity phenotypes. Methods: A total of 139 adults (31.00 ± 11.12 years) were classified into different metabolic phenotypes of obesity: 1) normal weight-lean (NWL group): BMI <25 kg/m2 and the fat-mass index (FMI) ≤9.7 kg/m2 in females and ≤6.3 kg/m2 in males; 2) metabolically obese normal weight (MONW group): BMI <25 kg/m2 and FMI >9.7 kg/m2 in females and >6.3 kg/m2 in males; 3) metabolically healthy obese (MHO group): BMI ≥25 and FMI ≤9.7 kg/m2 in females and ≤6.3 kg/m2 in males; and 4) metabolically unhealthy obese (MUO group): BMI ≥25 and FMI >9.7 kg/m2 in females and >6.3 kg/m2 in males. The GMA was measured at the baseline and post-prandial state using a multichannel electrogastrography with a water load satiety test. The average power distribution by the frequency region and the average dominant frequency were used for analysis. Anthropometric measurements and bioelectric impedance analysis were performed to calculate the FMI and fat-free mass index (FFMI). Serum levels of ghrelin, gastrin, and irisin were measured by ELISA kits according to the manufacturer’s protocol. Results: Compared to the NWL group, gastrin and ghrelin levels were significantly low in the MUO participants, while irisin was significantly high. The EGG showed significantly lower baseline and 20-min normogastria frequencies in the MHO and MUO groups. In the MHO group, baseline duodenal frequency was positively correlated with the gastrin level, while normogastria times were positively associated with the irisin level and negatively associated with the ghrelin level. In the MUO group, percentages of bradygastria frequencies at 10, 20, and 30 min were positively correlated with the BMI and FFMI. This bradygastria was correlated positively with the irisin level and negatively with the ghrelin level. Conclusion: The EGG patterns might be associated with obesity-related gastric hormones in different obesity phenotypes. EGG may be a promising clinical tool in obesity assessment. The association of the EGG patterns with hormonal levels needs further investigation for potential practical uses.
Collapse
Affiliation(s)
- Mahmoud M A Abulmeaty
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.,Obesity Management Unit, Medical Physiology Department, School of Medicine, Zagazig University, Zagazig, Egypt
| | - Dara Aldisi
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ghadeer S Aljuraiban
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ali Almajwal
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Eman El Shorbagy
- Obesity Management Unit, Medical Physiology Department, School of Medicine, Zagazig University, Zagazig, Egypt
| | - Yara Almuhtadi
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Batool Albaran
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Zaid Aldossari
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Thamer Alsager
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Razak
- Community Health Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Berika
- Rehabilitation Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed Al Zaben
- Surgery Department, Sultan Bin Abdulaziz Humanitarian City, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Cowan LM, Strege PR, Rusinova R, Andersen OS, Farrugia G, Beyder A. Capsaicin as an amphipathic modulator of Na V1.5 mechanosensitivity. Channels (Austin) 2022; 16:9-26. [PMID: 35412435 PMCID: PMC9009938 DOI: 10.1080/19336950.2022.2026015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
SCN5A-encoded NaV1.5 is a voltage-gated Na+ channel that drives the electrical excitability of cardiac myocytes and contributes to slow waves of the human gastrointestinal smooth muscle cells. NaV1.5 is mechanosensitive: mechanical force modulates several facets of NaV1.5’s voltage-gated function, and some NaV1.5 channelopathies are associated with abnormal NaV1.5 mechanosensitivity (MS). A class of membrane-active drugs, known as amphiphiles, therapeutically target NaV1.5’s voltage-gated function and produce off-target effects including alteration of MS. Amphiphiles may provide a novel option for therapeutic modulation of NaV1.5’s mechanosensitive operation. To more selectively target NaV1.5 MS, we searched for a membrane-partitioning amphipathic agent that would inhibit MS with minimal closed-state inhibition of voltage-gated currents. Among the amphiphiles tested, we selected capsaicin for further study. We used two methods to assess the effects of capsaicin on NaV1.5 MS: (1) membrane suction in cell-attached macroscopic patches and (2) fluid shear stress on whole cells. We tested the effect of capsaicin on NaV1.5 MS by examining macro-patch and whole-cell Na+ current parameters with and without force. Capsaicin abolished the pressure- and shear-mediated peak current increase and acceleration; and the mechanosensitive shifts in the voltage-dependence of activation (shear) and inactivation (pressure and shear). Exploring the recovery from inactivation and use-dependent entry into inactivation, we found divergent stimulus-dependent effects that could potentiate or mitigate the effect of capsaicin, suggesting that mechanical stimuli may differentially modulate NaV1.5 MS. We conclude that selective modulation of NaV1.5 MS makes capsaicin a promising candidate for therapeutic interventions targeting MS.
Collapse
Affiliation(s)
- Luke M Cowan
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Peter R Strege
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Radda Rusinova
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Olaf S Andersen
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Arthur Beyder
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| |
Collapse
|