1
|
Karimi P, Sunil M, Leong R, Ramirez-GarciaLuna JL, Ratcliffe E, Zuniga-Villanueva G. Prevalence of Gastrointestinal Adverse Events of Therapeutic Cannabinoids in Children: A Systematic Review and Meta-Analysis. Cannabis Cannabinoid Res 2025. [PMID: 39967452 DOI: 10.1089/can.2024.0104] [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: 02/20/2025] Open
Abstract
Introduction: The endocannabinoid system plays a crucial role in gastrointestinal homeostasis; although some gastrointestinal adverse events have been reported with therapeutic cannabinoids in children, the complete profile of gastrointestinal adverse events in the pediatric population remains unknown. Through a systematic review and meta-analysis, we aimed to identify the prevalence of gastrointestinal adverse events from therapeutic cannabinoids in children. Materials and Methods: A literature search of OVID MEDLINE, EMBASE, CINAHL, Web of Science, and The Cochrane Library was performed from inception to May 19, 2023. Selected studies included randomized controlled trials, retrospective cohort studies, uncontrolled before-after studies, and observational retrospective studies in English, French, or Spanish that reported gastrointestinal adverse events in the pediatric population under therapeutic cannabinoid interventions. The study was registered with PROSPERO and followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses reporting guidelines. A random-effects model was used to pool and analyze the extracted data. Extracted data included the presence of adverse gastrointestinal events by analyzing the type of cannabinoid, duration of treatment, dosage, and type of study. A subgroup meta-analysis was also performed, focusing on patients' conditions. Results: Twenty-five studies were included, comprising 1,201 pediatric patients receiving therapeutic cannabinoids, of whom 451 experienced gastrointestinal adverse events, representing a cumulative prevalence of 33.91% (95% confidence interval [CI]: 21.49% to 49.04%). Interventional studies reported a higher prevalence of GI adverse events (47.36%; 95% CI: 31% to 64%) compared with observational studies (17.6%; 95% CI: 8.5% to 32.7%). As most studies focused on patients with epilepsy, a subanalysis was performed within this population, revealing that patients with Dravet syndrome had a higher prevalence of diarrhea compared with other types of epilepsy (21.75%; 95% CI: 8.52% to 45.34% vs. 5.95%; 95% CI: 3.11% to 11.1%). Discussion: This systematic review and meta-analysis showed a high prevalence of gastrointestinal adverse events in children receiving therapeutic cannabinoids, with some populations, such as those with Dravet syndrome, being at higher risk than others. With the increased public discourse of cannabinoids being "natural" and mistakenly equating them as "risk-free," this information can help clinicians educate patients and the broader public on the adverse effects profile of these treatments.
Collapse
Affiliation(s)
- Parsa Karimi
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Maria Sunil
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Russell Leong
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | | | - Gregorio Zuniga-Villanueva
- Department of Pediatrics, McMaster University, Hamilton, Canada
- Department of Pediatrics, Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
2
|
Evans W, Akyea RK, Simms A, Kai J, Qureshi N. Opportunities and challenges for identifying undiagnosed Rare Disease patients through analysis of primary care records: long QT syndrome as a test case. J Community Genet 2024; 15:687-698. [PMID: 39405009 PMCID: PMC11645366 DOI: 10.1007/s12687-024-00742-7] [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: 03/28/2024] [Accepted: 10/02/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Patients with rare genetic diseases frequently experience significant diagnostic delays. Routinely collected data in the electronic health record (EHR) may be used to help identify patients at risk of undiagnosed conditions. Long QT syndrome (LQTS) is a rare inherited cardiac condition associated with significant morbidity and premature mortality. In this study, we examine LQTS as an exemplar disease to assess if clinical features recorded in the primary care EHR can be used to develop and validate a predictive model to aid earlier detection. METHODS 1495 patients with an LQTS diagnostic code and 7475 propensity-score matched controls were identified from 10.5 million patients' electronic primary care records in the UK's Clinical Practice Research Datalink (CPRD). Associated clinical features recorded before diagnosis (with p < 0.05) were incorporated into a multivariable logistic regression model, the final model was determined by backwards regression and validated by bootstrapping to determine model optimism. RESULTS The mean age at LQTS diagnosis was 58.4 (SD 19.41). 18 features were included in the final model. Discriminative accuracy, assessed by area under the curve (AUC), was 0.74, (95% CI 0.73, 0.75) (optimism 6%). Features occurring at significantly greater frequency before diagnosis included: epilepsy, palpitations, syncope, collapse, mitral valve disease and irritable bowel syndrome. CONCLUSION This study demonstrates the potential to develop primary care prediction models for rare conditions, like LQTS, in routine primary care records and highlights key considerations including disease suitability, finding an appropriate linked dataset, the need for accurate case ascertainment and utilising an approach to modelling suitable for rare events.
Collapse
Affiliation(s)
- William Evans
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK.
| | - Ralph K Akyea
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK
| | - Alex Simms
- Department of Cardiology, Leeds Teaching Hospital NHS Trust, Leeds, UK
| | - Joe Kai
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK
| | - Nadeem Qureshi
- Primary Care Stratified Medicine (PRISM), Centre for Academic Primary Care, School of Medicine, University of Nottingham, Applied Health Research Building [42], University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
3
|
Wang L, Tian H, Li L, Ye C, Cui J, Lin Z, Yang B, Zhao D, Li N, Feng X, Chen Q. To explore the postoperative nutritional status and factors influencing prognosis in patients with chronic constipation complicated by malnutrition. Front Med (Lausanne) 2024; 11:1335203. [PMID: 39290393 PMCID: PMC11405215 DOI: 10.3389/fmed.2024.1335203] [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: 11/08/2023] [Accepted: 08/22/2024] [Indexed: 09/19/2024] Open
Abstract
Background Many patients with constipation also suffer from varying degrees of malnutrition, and the relationship between the two conditions is a vicious cycle. Surgery is the final step in the treatment of constipation, with a success rate of up to 95%. This study aims to investigate the effects of surgical treatment on the nutritional status of patients with chronic constipation and malnutrition. Methods A total of 60 patients with chronic constipation and various degrees of malnutrition who underwent surgery in our department from January 2020 to March 2023 were included in this study. Biochemical tests including BMI, albumin, total protein, hemoglobin, cholesterol and lymphocyte count were conducted, as well as measurements of inflammatory markers such as Interleukin-6 (IL-6), Interleukin-8 (IL-8), and C-reactive protein (CRP). Additionally, multiple nutritional risk screening scales (NRS2002, MUST, NRI, and MNA) and the prognostic nutritional index (PNI) were used to assess the nutritional status of patients before surgery, as well as at 1 month, 3 months, and 6 months post-surgery. Finally, we analyzed the factors influencing postoperative recovery outcomes in patients. Results Compared to pre-operation, the BMI of patients significantly increased at 1 month, 3 months, and 6 months after the operation, with statistically significant differences (p < 0.001). Multiple nutritional risk assessment tools (NRS2002, MUST, NRI, and MNA), as well as the prognostic nutritional index (NPI), indicated a reduction in nutritional risk and improvement in nutritional status at 1, 3, and 6 months post-surgery, compared to pre-surgery levels (p < 0.001). The levels of albumin, total protein, and hemoglobin in patients at 1, 3, and 6 months after the surgery were significantly higher than those before the surgery (p < 0.001). However, there was no significant change in the number of lymphocytes. Inflammatory markers such as IL-6, IL-8, and CRP exhibited a significant decrease after the surgery, reaching normal levels at 6 months post-surgery (p < 0.001). Low BMI, low PNI, and low cholesterol levels are independent risk factors for patient prognosis (p < 0.05). Conclusion Surgical treatment can enhance the nutritional status of constipation patients with malnutrition, which in turn promotes the restoration of intestinal motility. The patient's nutritional status will impact the postoperative recovery outcomes.
Collapse
Affiliation(s)
- Le Wang
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Hongliang Tian
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
- Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai, China
| | - Long Li
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Chen Ye
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Jiaqu Cui
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Zhiliang Lin
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Bo Yang
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Di Zhao
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Ning Li
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
| | - Xiaobo Feng
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Qiyi Chen
- Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Gastrointestinal Microecology Research Center, Shanghai, China
- Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai, China
| |
Collapse
|
4
|
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
|
5
|
Maqoud F, Tricarico D, Mallamaci R, Orlando A, Russo F. The Role of Ion Channels in Functional Gastrointestinal Disorders (FGID): Evidence of Channelopathies and Potential Avenues for Future Research and Therapeutic Targets. Int J Mol Sci 2023; 24:11074. [PMID: 37446251 DOI: 10.3390/ijms241311074] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Several gastrointestinal (GI) tract abnormalities, including visceral hypersensitivity, motility, and intestinal permeability alterations, have been implicated in functional GI disorders (FGIDs). Ion channels play a crucial role in all the functions mentioned above. Hormones and natural molecules modulate these channels and represent targets of drugs and bacterial toxins. Mutations and abnormal functional expression of ion channel subunits can lead to diseases called channelopathies. These channelopathies in gastroenterology are gaining a strong interest, and the evidence of co-relationships is increasing. In this review, we describe the correlation status between channelopathies and FGIDs. Different findings are available. Among others, mutations in the ABCC7/CFTR gene have been described as a cause of constipation and diarrhea. Mutations of the SCN5A gene are instead associated with irritable bowel syndrome. In contrast, mutations of the TRPV1 and TRPA genes of the transient receptor potential (TRP) superfamily manifest hypersensitivity and visceral pain in sensory nerves. Recently, mice and humans affected by Cantu syndrome (CS), which is associated with the mutations of the KCNJ8 and ABCC9 genes encoding for the Kir6.1 and SUR2 subunits, showed dysfunction of contractility throughout the intestine and death in the mice after the weaning on solid food. The discovery of a correlation between channelopathies and FIGD opens new avenues for discovering new direct drug targets for specific channelopathies, leading to significant implications for diagnosing and treating functional GI diseases.
Collapse
Affiliation(s)
- Fatima Maqoud
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Castellana Grotte, 70013 Bari, Italy
| | - Domenico Tricarico
- Section of Pharmacology, Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Rosanna Mallamaci
- Department of Biosciences, Biotechnologies and Environment University of Bari Aldo Moro, 70125 Bari, Italy
| | - Antonella Orlando
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Castellana Grotte, 70013 Bari, Italy
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS "Saverio de Bellis", Castellana Grotte, 70013 Bari, Italy
| |
Collapse
|
6
|
Carlos dos Reis D, Dastoor P, Santos AK, Sumigray K, Ameen NA. CFTR high expresser cells in cystic fibrosis and intestinal diseases. Heliyon 2023; 9:e14568. [PMID: 36967909 PMCID: PMC10031467 DOI: 10.1016/j.heliyon.2023.e14568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), the Cl-/HCO3 - channel implicated in Cystic Fibrosis, is critical to the pathophysiology of many gastrointestinal diseases. Defects in CFTR lead to intestinal dysfunction, malabsorption, obstruction, infection, inflammation, and cancer that increases morbidity and reduces quality of life. This review will focus on CFTR in the intestine and the implications of the subpopulation of CFTR High Expresser Cells (CHEs) in Cystic Fibrosis (CF), intestinal physiology and pathophysiology of intestinal diseases.
Collapse
Affiliation(s)
- Diego Carlos dos Reis
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
| | - Parinaz Dastoor
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
| | - Anderson Kenedy Santos
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Kaelyn Sumigray
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, 06510, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Nadia A. Ameen
- Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT06510, USA
- Corresponding author. Department of Pediatrics/Gastroenterology and Hepatology, Yale School of Medicine, CT, 06510, USA.
| |
Collapse
|
7
|
Xie Z, Feng J, Hibberd TJ, Chen BN, Zhao Y, Zang K, Hu X, Yang X, Chen L, Brookes SJ, Spencer NJ, Hu H. Piezo2 channels expressed by colon-innervating TRPV1-lineage neurons mediate visceral mechanical hypersensitivity. Neuron 2023; 111:526-538.e4. [PMID: 36563677 PMCID: PMC9957938 DOI: 10.1016/j.neuron.2022.11.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/13/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022]
Abstract
Inflammatory and functional gastrointestinal disorders such as irritable bowel syndrome (IBS) and obstructive bowel disorder (OBD) underlie the most prevalent forms of visceral pain. Although visceral pain can be generally provoked by mechanical distension/stretch, the mechanisms that underlie visceral mechanosensitivity in colon-innervating visceral afferents remain elusive. Here, we show that virally mediated ablation of colon-innervating TRPV1-expressing nociceptors markedly reduces colorectal distention (CRD)-evoked visceromotor response (VMR) in mice. Selective ablation of the stretch-activated Piezo2 channels from TRPV1 lineage neurons substantially reduces mechanically evoked visceral afferent action potential firing and CRD-induced VMR under physiological conditions, as well as in mouse models of zymosan-induced IBS and partial colon obstruction (PCO). Collectively, our results demonstrate that mechanosensitive Piezo2 channels expressed by TRPV1-lineage nociceptors powerfully contribute to visceral mechanosensitivity and nociception under physiological conditions and visceral hypersensitivity under pathological conditions in mice, uncovering potential therapeutic targets for the treatment of visceral pain.
Collapse
Affiliation(s)
- Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA; Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Bao Nan Chen
- Neurogastroenterology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Yonghui Zhao
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Kaikai Zang
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Xueming Hu
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Xingliang Yang
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Lvyi Chen
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA; School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, Hubei, People's Republic of China
| | - Simon J Brookes
- Neurogastroenterology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia.
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
8
|
Li Y, Yu H, Xu B, Yuan F, Zhang P, Tu S, Zhou Y, Li X. Pharmacokinetics and Bioequivalence of the Lubiprostone Capsule in Healthy Chinese Subjects. Clin Pharmacol Drug Dev 2023; 12:436-446. [PMID: 36626291 DOI: 10.1002/cpdd.1212] [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: 09/20/2022] [Accepted: 11/29/2022] [Indexed: 01/11/2023]
Abstract
A pharmacokinetic (PK) study and a bioequivalence (BE) study were conducted to investigate the PK characteristics and safety of lubiprostone in healthy Chinese subjects and to evaluate the BE between the test and the reference drugs. The PK study consisted of a fasting state cohort (a single dose of 24 µg of lubiprostone), a 2-period crossover fasting and fed state cohort (a single dose of 48 µg of lubiprostone), and a multiple-dose cohort (24 µg of lubiprostone twice daily). The BE study was a single-dose, 2-treatment, 4-period, replicated crossover study. The plasma concentration of 15-OH-lubiprostone (M3) was measured by high-performance liquid chromatography-tandem mass spectrometry. The PK parameters were calculated using the noncompartment model with Phoenix WinNonlin. After a single dose of 24 ug of lubiprostone, the main PK parameters of M3 were 49.2 pg/mL, 74.0 h/pg/mL, and 1.1 hours for maximum plasma concentration (Cmax ), area under the plasma concentration time curve from time 0 to the last time point, and t1/2 , respectively. The main PK parameters of M3 showed dose-proportional characteristics in the dose range of 24-48 µg. Food affects the PK parameters of M3. Compared to the fasting state, time to maximum plasma concentration was delayed, Cmax decreased slightly, while AUC increased significantly under the fed state. The test and reference products had similar PK parameters and were bioequivalent in the fed state.
Collapse
Affiliation(s)
- Yuan Li
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Haitao Yu
- Nanjing Chia Tai Tianqing Pharmaceutical Co., Ltd., Nanjing, Jiangsu, China
| | - Bing Xu
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Fang Yuan
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Ping Zhang
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Shengqing Tu
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Yumeng Zhou
- Nanjing Chia Tai Tianqing Pharmaceutical Co., Ltd., Nanjing, Jiangsu, China
| | - Xin Li
- Department of Pharmacy, The Third Hospital of Changsha, Changsha, Hunan, China
| |
Collapse
|
9
|
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
|
10
|
Feng J, Xie Z, Hu H. Ion channel regulation of gut immunity. J Gen Physiol 2022; 155:213734. [PMID: 36459135 PMCID: PMC9723512 DOI: 10.1085/jgp.202113042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/15/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Mounting evidence indicates that gastrointestinal (GI) homeostasis hinges on communications among many cellular networks including the intestinal epithelium, the immune system, and both intrinsic and extrinsic nerves innervating the gut. The GI tract, especially the colon, is the home base for gut microbiome which dynamically regulates immune function. The gut's immune system also provides an effective defense against harmful pathogens entering the GI tract while maintaining immune homeostasis to avoid exaggerated immune reaction to innocuous food and commensal antigens which are important causes of inflammatory disorders such as coeliac disease and inflammatory bowel diseases (IBD). Various ion channels have been detected in multiple cell types throughout the GI tract. By regulating membrane properties and intracellular biochemical signaling, ion channels play a critical role in synchronized signaling among diverse cellular components in the gut that orchestrates the GI immune response. This work focuses on the role of ion channels in immune cells, non-immune resident cells, and neuroimmune interactions in the gut at the steady state and pathological conditions. Understanding the cellular and molecular basis of ion channel signaling in these immune-related pathways and initial testing of pharmacological intervention will facilitate the development of ion channel-based therapeutic approaches for the treatment of intestinal inflammation.
Collapse
Affiliation(s)
- Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO,Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, China,Correspondence to Jing Feng:
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch and Sensory Disorders, Washington University School of Medicine, St. Louis, MO,Hongzhen Hu:
| |
Collapse
|
11
|
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: 0.3] [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
|
12
|
Sadraei H, Ghasemi M, Saranji S. Evaluation of spasmolytic effects of naringenin on ileum contraction and intestinal charcoal meal transit: Involvement of ATP-sensitive K+ channels. JOURNAL OF HERBMED PHARMACOLOGY 2022. [DOI: 10.34172/jhp.2022.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Naringenin is a flavonoid constituent of many herbal plants, including citreous fruits. Biological studies have suggested various therapeutic effects for naringenin, including protective effects on gastrointestinal (GI) motility. The present study was performed to investigate the involvement of ATP-sensitive K+ channels on the effect of naringenin in rat ileum motility. Methods: Ileum contractions were induced by either KCl or acetylcholine (ACh) in vitro. Inhibitory concentration-response curves were constructed for naringenin and diazoxide after exposure of rat isolated ileum to KCl (20mM) or ACh (500nM). The relaxant effects of naringenin and diazoxide were also examined in the presence of glibenclamide. Furthermore, oral effects of diazoxide (25 mg/kg) and naringenin (25, 50 mg/kg) were also assessed on the intestinal charcoal meal transit in mice (n=10) in the absence and presence of glibenclamide (50 mg/kg). Results: Diazoxide and naringenin in a concentration-dependent manner inhibited ileum contractions induced by low bath concentration of KCl (20mM). However, both drugs had no effect on contractions induced by a high concentration of KCl (160mM). The inhibitory effects of diazoxide and naringenin were blocked by glibenclamide. Oral administration of diazoxide and naringenin significantly reduced the intestinal transit of charcoal meal. The delay in the intestinal transit was blocked by the oral dose of glibenclamide. The effect of naringenin on the rat intestinal strip pre-contracted with the KCl was relatively similar to that of ATP-sensitive K+ channel opener (diazoxide). Conclusion: This research supports that ATP-sensitive K+ channels are involved in the rat small intestinal smooth muscles relaxation induced by naringenin.
Collapse
Affiliation(s)
- Hassan Sadraei
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maedeh Ghasemi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Salman Saranji
- Department of Pharmacology and Toxicology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
13
|
Chan CHA, Aghababaie Z, Paskaranandavadivel N, Avci R, Cheng LK, Angeli-Gordon TR. Localized gastric distension disrupts slow-wave entrainment leading to temporary ectopic propagation: a high-resolution electrical mapping study. Am J Physiol Gastrointest Liver Physiol 2021; 321:G656-G667. [PMID: 34612062 DOI: 10.1152/ajpgi.00219.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric distension is known to affect normal slow-wave activity and gastric function, but links between slow-wave dysrhythmias and stomach function are poorly understood. Low-resolution mapping is unable to capture complex spatial properties of gastric dysrhythmias, necessitating the use of high-resolution mapping techniques. Characterizing the nature of these dysrhythmias has implications in the understanding of postprandial function and the development of new mapping devices. In this two-phase study, we developed and implemented a protocol for measuring electrophysiological responses to gastric distension in porcine experiments. In vivo, serosal high-resolution electrical mapping (256 electrodes; 36 cm2) was performed in anaesthetized pigs (n = 11), and slow-wave pattern, velocity, frequency, and amplitude were quantified before, during, and after intragastric distension. Phase I experiments (n = 6) focused on developing and refining the distension mapping methods using a surgically inserted intragastric balloon, with a variety of balloon types and distension protocols. Phase II experiments (n = 5) used barostat-controlled 500-mL isovolumetric distensions of an endoscopically introduced intragastric balloon. Dysrhythmias were consistently induced in all five gastric distensions, using refined distension protocols. Dysrhythmias appeared 23 s (SD = 5 s) after the distension and lasted 129 s (SD = 72 s), which consisted of ectopic propagation originating from the greater curvature in the region of distension. In summary, our results suggest that distension disrupts gastric entrainment, inducing temporary ectopic slow-wave propagation. These results may influence the understanding of the postprandial stomach and electrophysiological effects of gastric interventions.NEW & NOTEWORTHY This study presents the discovery of temporary dysrhythmic ectopic pacemakers in the distal stomach caused by localized gastric distension. Distension-induced dysrhythmias are an interesting physiological phenomenon that can inform the design of new interventional and electrophysiological protocols for both research and the clinic. The observation of distension-induced dysrhythmias also contributes to our understanding of stretch-sensitivity in the gut and may play an important role in normal and abnormal postprandial physiology.
Collapse
Affiliation(s)
| | - Zahra Aghababaie
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Niranchan Paskaranandavadivel
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Recep Avci
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Leo K Cheng
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Department of Surgery, Vanderbilt University, Nashville, Tennessee
| | - Timothy R Angeli-Gordon
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Department of Surgery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Sarica AS, Bor S, Orman MN, Barajas-Martinez H, Juang JMJ, Antzelevitch C, Hasdemir C. Frequency of Irritable Bowel Syndrome in Patients with Brugada Syndrome and Drug-Induced Type 1 Brugada Pattern. Am J Cardiol 2021; 151:51-56. [PMID: 34034907 DOI: 10.1016/j.amjcard.2021.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Irritable bowel syndrome (IBS) is one of the most widely recognized functional bowel disorders (FBDs) with a genetic component. SCN5A gene and SCN1B loci have been identified in population-based IBS cohorts and proposed to have a mechanistic role in the pathophysiology of IBS. These same genes have been associated with Brugada syndrome (BrS). The present study examines the hypothesis that these two inherited syndromes are linked. Prevalence of FBDs over a 12 months period were compared between probands with BrS/drug-induced type 1 Brugada pattern (DI-Type 1 BrP) (n = 148) and a control group (n = 124) matched for age, female sex, presence of arrhythmia and co-morbid conditions. SCN5A/SCN1B genes were screened in 88 patients. Prevalence of IBS was 25% in patients with BrS/DI-Type 1 BrP and 8.1% in the control group (p = 2.34 × 10-4). On stepwise logistic regression analysis, presence of current and/or history of migraine (OR of 2.75; 95% CI: 1.08 to 6.98; p = 0.033) was a predictor of underlying BrS/DI-Type 1 BrP among patients with FBDs. We identified 8 putative SCN5A/SCN1B variants in 7 (12.3%) patients with BrS/DI-Type 1 BrP and 1 (3.2%) patient in control group. Five out of 8 (62.5%) patients with SCN5A/SCN1B variants had FBDs. In conclusion, IBS is a common co-morbidity in patients with BrS/DI-Type 1 BrP. Presence of current and/or history of migraine are a predictor of underlying BrS/DI-Type 1 BrP among patients with FBDs. Frequent co-existence of IBS and BrS/DI-Type 1 BrP necessitates cautious use of certain drugs among the therapeutic options for IBS that are known to exacerbate the Brugada phenotype.
Collapse
|
15
|
Nagahawatte ND, Paskaranandavadivel N, Angeli-Gordon TR, Cheng LK, Avci R. Transmural recordings of gastrointestinal electrical activity using a spatially-dense microelectrode array. Physiol Meas 2021; 42. [PMID: 33607644 DOI: 10.1088/1361-6579/abe80f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/19/2021] [Indexed: 11/11/2022]
Abstract
Objective. High-resolution serosal recordings provide detailed information about the bioelectrical conduction patterns in the gastrointestinal (GI) tract. However, equivalent knowledge about the electrical activity through the GI tract wall remains largely unknown. This study aims to capture and quantify the bioelectrical activity across the wall of the GI tract.Approach. A needle-based microelectrode array was used to measure the bioelectrical activity across the GI wallin vivo. Quantitative and qualitative evaluations of transmural slow wave characteristics were carried out in comparison to the serosal slow wave features, through which the period, amplitude, and SNR metrics were quantified and statistically compared.Main results. Identical periods of 4.7 ± 0.3 s with amplitudes of 0.17 ± 0.04 mV versus 0.31 ± 0.1 mV and signal to noise ratios of 5.5 ± 1.3 dB versus 14.4 ± 1.1 dB were observed for transmural and serosal layers, respectively. Four different slow wave morphologies were observed across the transmural layers of the GI wall. Similar amplitudes were observed for all morphology types, and Type 1 and Type 2 were of the highest prevalence, dominating the outer and inner layers. Type 2 was exclusive to the middle layer while Type 4 was primarily observed in the middle layer as well.Significance. This study demonstrates the validity of new methodologies for measuring transmural slow wave activation in the GI wall and can now be applied to investigate the source and origin of GI dysrhythmias leading to dysmotility, and to validate novel therapeutics for GI health and disease.
Collapse
Affiliation(s)
- Nipuni D Nagahawatte
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | | | - Timothy R Angeli-Gordon
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Riddet Institute Centre of Research Excellence, Palmerston North, New Zealand
| | - Leo K Cheng
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand.,Riddet Institute Centre of Research Excellence, Palmerston North, New Zealand.,Department of Surgery, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Recep Avci
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Buddington RK, Wong T, Howard SC. Paracellular Filtration Secretion Driven by Mechanical Force Contributes to Small Intestinal Fluid Dynamics. Med Sci (Basel) 2021; 9:medsci9010009. [PMID: 33572202 PMCID: PMC7931054 DOI: 10.3390/medsci9010009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Studies of fluid secretion by the small intestine are dominated by the coupling with ATP-dependent generation of ion gradients, whereas the contribution of filtration secretion has been overlooked, possibly by the lack of a known mechanistic basis. We measured apical fluid flow and generation of hydrostatic pressure gradients by epithelia of cultured mouse enterocytes, Caco-2 and T-84 cells, and fibroblasts exposed to mechanical force provided by vigorous aeration and in response to ion gradients, inhibitors of ion channels and transporters and in vitro using intact mouse and rat small intestine. We describe herein a paracellular pathway for unidirectional filtration secretion that is driven by mechanical force, requires tight junctions, is independent of ionic and osmotic gradients, generates persistent hydrostatic pressure gradients, and would contribute to the fluid shifts that occur during digestion and diarrhea. Zinc inhibits the flow of fluid and the paracellular marker fluorescein isothyocyanate conjugated dextran (MW = 4 kD) across epithelia of cultured enterocytes (>95%; p < 0.001) and intact small intestine (>40%; p = 0.03). We propose that mechanical force drives fluid secretion through the tight junction complex via a “one-way check valve” that can be regulated. This pathway of filtration secretion complements chloride-coupled fluid secretion during high-volume fluid flow. The role of filtration secretion in the genesis of diarrhea in intact animals needs further study. Our findings may explain a potential linkage between intestinal motility and intestinal fluid dynamics.
Collapse
Affiliation(s)
- Randal K. Buddington
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA;
- Babies Taking Flight, Memphis, TN 38117, USA
- Correspondence: ; Tel.: +1-662-418-2666
| | - Thomas Wong
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA;
| | - Scott C. Howard
- Department of Acute and Tertiary Care, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA;
| |
Collapse
|
17
|
Chen A, Majdinasab EJ, Fiori MC, Liang H, Altenberg GA. Polymer-Encased Nanodiscs and Polymer Nanodiscs: New Platforms for Membrane Protein Research and Applications. Front Bioeng Biotechnol 2020; 8:598450. [PMID: 33304891 PMCID: PMC7701119 DOI: 10.3389/fbioe.2020.598450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
Membrane proteins (MPs) are essential to many organisms’ major functions. They are notorious for being difficult to isolate and study, and mimicking native conditions for studies in vitro has proved to be a challenge. Lipid nanodiscs are among the most promising platforms for MP reconstitution, but they contain a relatively labile lipid bilayer and their use requires previous protein solubilization in detergent. These limitations have led to the testing of copolymers in new types of nanodisc platforms. Polymer-encased nanodiscs and polymer nanodiscs support functional MPs and address some of the limitations present in other MP reconstitution platforms. In this review, we provide a summary of recent developments in the use of polymers in nanodiscs.
Collapse
Affiliation(s)
- Angela Chen
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Elleana J Majdinasab
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Mariana C Fiori
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hongjun Liang
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Guillermo A Altenberg
- Department of Cell Physiology and Molecular Biophysics, and Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
18
|
Klimovich A, Giacomello S, Björklund Å, Faure L, Kaucka M, Giez C, Murillo-Rincon AP, Matt AS, Willoweit-Ohl D, Crupi G, de Anda J, Wong GCL, D'Amato M, Adameyko I, Bosch TCG. Prototypical pacemaker neurons interact with the resident microbiota. Proc Natl Acad Sci U S A 2020; 117:17854-17863. [PMID: 32647059 PMCID: PMC7395494 DOI: 10.1073/pnas.1920469117] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pacemaker neurons exert control over neuronal circuit function by their intrinsic ability to generate rhythmic bursts of action potential. Recent work has identified rhythmic gut contractions in human, mice, and hydra to be dependent on both neurons and the resident microbiota. However, little is known about the evolutionary origin of these neurons and their interaction with microbes. In this study, we identified and functionally characterized prototypical ANO/SCN/TRPM ion channel-expressing pacemaker cells in the basal metazoan Hydra by using a combination of single-cell transcriptomics, immunochemistry, and functional experiments. Unexpectedly, these prototypical pacemaker neurons express a rich set of immune-related genes mediating their interaction with the microbial environment. Furthermore, functional experiments gave a strong support to a model of the evolutionary emergence of pacemaker cells as neurons using components of innate immunity to interact with the microbial environment and ion channels to generate rhythmic contractions.
Collapse
Affiliation(s)
- Alexander Klimovich
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany;
| | - Stefania Giacomello
- Department of Biochemistry and Biophysics, National Infrastructure of Sweden, Science for Life Laboratory, Stockholm University, 17121 Solna, Sweden
- Department of Gene Technology, Science for Life Laboratory, Kungligia Tekniska Högskolan Royal Institute of Technology, 17121 Solna, Sweden
| | - Åsa Björklund
- Department of Cell and Molecular Biology, National Infrastructure of Sweden, Science for Life Laboratory, Uppsala University, 75237 Uppsala, Sweden
| | - Louis Faure
- Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
| | - Marketa Kaucka
- Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Biology, SH 24306 Plön, Germany
| | - Christoph Giez
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany
| | - Andrea P Murillo-Rincon
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany
| | - Ann-Sophie Matt
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany
| | - Doris Willoweit-Ohl
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany
| | - Gabriele Crupi
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany
| | - Jaime de Anda
- Department of Bioengineering, California NanoSystems Institute, University of California, Los Angeles, CA 90095-1600
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, CA 90095-1600
| | - Gerard C L Wong
- Department of Bioengineering, California NanoSystems Institute, University of California, Los Angeles, CA 90095-1600
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, CA 90095-1600
| | - Mauro D'Amato
- School of Biological Sciences, Monash University, Clayton, VIC 3800, Australia
| | - Igor Adameyko
- Department of Molecular Neurosciences, Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Thomas C G Bosch
- Department of Cell and Developmental Biology, Zoological Institute, University of Kiel, D-24118 Kiel, Germany;
| |
Collapse
|
19
|
TASK-3 Gene Knockdown Dampens Invasion and Migration and Promotes Apoptosis in KATO III and MKN-45 Human Gastric Adenocarcinoma Cell Lines. Int J Mol Sci 2019; 20:ijms20236077. [PMID: 31810225 PMCID: PMC6928893 DOI: 10.3390/ijms20236077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
Incidence and mortality of gastric cancer is increasing worldwide, in part, because of the lack of new therapeutic targets to treat this disease. Different types of ion channels participate in the hallmarks of cancer. In this context, ion channels are known to exert control over the cell cycle, mechanisms that support survival, angiogenesis, migration, and cell invasion. In particular, TASK-3 (KCNK9), a member of the K2P potassium channel family, has attracted much interest because of its oncogenic properties. However, despite multiple lines of evidence linking TASK-3 to tumorigenesis in various types of cancer, its relationship with gastric cancer has not been fully examined. Therefore, we set out to assess the effect of TASK-3 gene knockdown on KATO III and MKN-45 human gastric adenocarcinoma cell lines by using a short hairpin RNA (shRNA)-mediated knockdown. Our results demonstrate that knocking down TASK-3 reduces cell proliferation and viability because of an increase in apoptosis without an apparent effect on cell cycle checkpoints. In addition, cell migration and invasion are reduced after knocking down TASK-3 in these cell lines. The present study highlights TASK-3 as a key protein involved in migration and cell survival in gastric cancer and corroborates its potential as a therapeutic target for gastric cancer treatment.
Collapse
|
20
|
Yeh KM, Johansson O, Le H, Rao K, Markus I, Perera DS, Lubowski DZ, King DW, Zhang L, Chen H, Liu L. Cystic fibrosis transmembrane conductance regulator modulates enteric cholinergic activities and is abnormally expressed in the enteric ganglia of patients with slow transit constipation. J Gastroenterol 2019; 54:994-1006. [PMID: 31392489 DOI: 10.1007/s00535-019-01610-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/31/2019] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cystic fibrosis transmembrane conductance regulator (CFTR) was recently found in the enteric nervous system, where its role is unclear. We aimed to identify which enteric neuronal structures express CFTR, whether CFTR modulates enteric neurotransmission and if altered CFTR expression is associated with slow transit constipation (STC). METHODS Immunofluorescence double labeling was performed to localize CFTR with various neuronal and glial cell markers in the human colon. The immunoreactivity (IR) of CFTR and choline acetyltransferase (ChAT) on myenteric plexus of control and STC colon was quantitatively analyzed. In control colonic muscle strips, electrical field stimulation (EFS) evoked contractile responses and the release of acetylcholine (ACh) was measured in the presence of the CFTR channel inhibitor, CFTR(inh)-172. RESULTS CFTR-IR was densely localized to myenteric ganglia, where it was co-localized with neuronal markers HuC/D and β-tubulin, and glial marker S-100 but little with glial fibrillary acidic protein. Vesicular ACh transport was almost exclusively co-localized with CFTR, but neurons expressing nitric oxide synthase were CFTR negative. Significant reductions of CFTR-IR (P < 0.01) and ChAT-IR (P < 0.05) were observed on myenteric ganglia of STC compared to control. Pre-treatment of colonic muscle strips with CFTR(inh)-172 (10 µM) significantly inhibited EFS-evoked contractile responses (P < 0.01) and ACh release (P < 0.05). CONCLUSIONS Co-localization of CFTR-IR with cholinergic markers, inhibition of EFS-induced colonic muscle contractility and ACh release by CFTR(inh)-172 suggest that CFTR modulates enteric cholinergic neurotransmission. The downregulation of CFTR and ChAT in myenteric ganglia of STC correlated with the impaired contractile responses to EFS.
Collapse
Affiliation(s)
- Ka Ming Yeh
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Olle Johansson
- Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Huy Le
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Karan Rao
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Irit Markus
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | | | | | | | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Hongzhuan Chen
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lu Liu
- Department of Pharmacology, Faculty of Medicine, School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
21
|
Marine Toxins and Nociception: Potential Therapeutic Use in the Treatment of Visceral Pain Associated with Gastrointestinal Disorders. Toxins (Basel) 2019; 11:toxins11080449. [PMID: 31370176 PMCID: PMC6723473 DOI: 10.3390/toxins11080449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Visceral pain, of which the pathogenic basis is currently largely unknown, is a hallmark symptom of both functional disorders, such as irritable bowel syndrome, and inflammatory bowel disease. Intrinsic sensory neurons in the enteric nervous system and afferent sensory neurons of the dorsal root ganglia, connecting with the central nervous system, represent the primary neuronal pathways transducing gut visceral pain. Current pharmacological therapies have several limitations, owing to their partial efficacy and the generation of severe adverse effects. Numerous cellular targets of visceral nociception have been recognized, including, among others, channels (i.e., voltage-gated sodium channels, VGSCs, voltage-gated calcium channels, VGCCs, Transient Receptor Potential, TRP, and Acid-sensing ion channels, ASICs) and neurotransmitter pathways (i.e., GABAergic pathways), which represent attractive targets for the discovery of novel drugs. Natural biologically active compounds, such as marine toxins, able to bind with high affinity and selectivity to different visceral pain molecular mediators, may represent a useful tool (1) to improve our knowledge of the physiological and pathological relevance of each nociceptive target, and (2) to discover therapeutically valuable molecules. In this review we report the most recent literature describing the effects of marine toxin on gastrointestinal visceral pain pathways and the possible clinical implications in the treatment of chronic pain associated with gut diseases.
Collapse
|
22
|
Li B, Rui J, Ding X, Yang X. Exploring the multicomponent synergy mechanism of Banxia Xiexin Decoction on irritable bowel syndrome by a systems pharmacology strategy. JOURNAL OF ETHNOPHARMACOLOGY 2019; 233:158-168. [PMID: 30590198 DOI: 10.1016/j.jep.2018.12.033] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/29/2018] [Accepted: 12/20/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin Decoction (BXD) is a representative prescription to regulate spleen and stomach in "Treatise on Febrile Diseases", which has been proven effective for the clinical treatment of irritable bowel syndrome (IBS) in the past decades. However, the active principles and molecular mechanisms involved in BXD against IBS are vague yet. AIM OF THE STUDY To unfold multicomponent synergy mechanism of BXD on irritable bowel syndrome, this work explored active principles, drug targets and crucial pathways using a systems pharmacology strategy. MATERIALS AND METHODS In this study, a systems pharmacology based strategy was applied by the procedures integrating compound database construction, ADME evaluation, target identification, functional annotation, pathway enrichment analysis, network analysis, and molecular docking verification. The 158 compounds from BXD were selected for the screening. The Compound-Target network (C-T) and the Target-Pathway network (T-P) were constructed. The bioinformatics and network topology were employed to systematically reveal multicomponent-target interactions of BXD. The affinity between important ingredients and the kernel targets was validated using molecular mechanics simulation. RESULTS The 35 potential important ingredients and the 16 associated kernel targets were identified. 27 crucial pathways, in which the kernel targets participated, could regulate the biological processes, such as synthesis of inflammatory mediators, smooth muscle relaxation and synaptic plasticity, closely related to pathological mechanism of IBS. The cross-talk interactions were revealed between TNF signaling pathway, Dopaminergic synapse and cGMP-PKG signaling pathway, which would exert the synergistic influences on the occurrence and treatment of the IBS. PTGS2, CALM, NOS2, SCN5A, and PRSS1 might become novel drug targets for IBS. CONCLUSIONS The study demonstrated that the synergy molecular mechanisms of BXD mainly involved three therapeutic modules including inhibiting inflammatory reaction, maintaining intestinal function and improving psychological regulation via the multicomponent-target interaction networks. It may also provide the promising drug targets and therapeutic agents for the development of new medicines.
Collapse
Affiliation(s)
- Bangjie Li
- College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Junqian Rui
- College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Xuejian Ding
- College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Xinghao Yang
- College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
23
|
Picard E, Carvalho FA, Agosti F, Bourinet E, Ardid D, Eschalier A, Daulhac L, Mallet C. Inhibition of Ca v 3.2 calcium channels: A new target for colonic hypersensitivity associated with low-grade inflammation. Br J Pharmacol 2019; 176:950-963. [PMID: 30714145 DOI: 10.1111/bph.14608] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/13/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Abdominal pain associated with low-grade inflammation is frequently encountered in irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD) during remission. Current treatments are not very effective and new therapeutic approaches are needed. The role of CaV 3.2 channels, which are important in other chronic pain contexts, was investigated in a murine model of colonic hypersensitivity (CHS) associated with low-grade inflammation. EXPERIMENTAL APPROACH Low doses of dextran sulfate sodium (DSS; 0.5%) were chronically administered to C57BL/6j mice in drinking water. Their inflammatory state was assessed by systemic and local measures of IL-6, myeloperoxidase, and lipocalin-2 using elisa. Colonic sensitivity was evaluated by the visceromotor responses to colorectal distension. Functional involvement of CaV 3.2 channels was assessed with different pharmacological (TTA-A2, ABT-639, and ethosuximide) and genetic tools. KEY RESULTS DSS induced low-grade inflammation associated with CHS in mice. Genetic or pharmacological inhibition of CaV 3.2 channels reduced CHS. Cav3.2 channel deletion in primary nociceptive neurons in dorsal root ganglia (CaV 3.2Nav1.8 KO mice) suppressed CHS. Spinal, but not systemic, administration of ABT-639, a peripherally acting T-type channel blocker, reduced CHS. ABT-639 given intrathecally to CaV 3.2Nav1.8 KO mice had no effect, demonstrating involvement of CaV 3.2 channels located presynaptically in afferent fibre terminals. Finally, ethosuximide, which is a T-type channel blocker used clinically, reduced CHS. CONCLUSIONS AND IMPLICATIONS These results suggest that ethosuximide represents a promising drug reposition strategy and that inhibition of CaV 3.2 channels is an attractive therapeutic approach for relieving CHS in IBS or IBD.
Collapse
Affiliation(s)
- Elodie Picard
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Frederic Antonio Carvalho
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Francina Agosti
- CNRS, INSERM, LABEX ICST, IGF, Université Montpellier, Montpellier, France
| | - Emmanuel Bourinet
- CNRS, INSERM, LABEX ICST, IGF, Université Montpellier, Montpellier, France
| | - Denis Ardid
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Laurence Daulhac
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Christophe Mallet
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| |
Collapse
|
24
|
Masotti M, Delprete C, Dothel G, Donadio V, Rimondini R, Politei JM, Liguori R, Caprini M. Altered globotriaosylceramide accumulation and mucosal neuronal fiber density in the colon of the Fabry disease mouse model. Neurogastroenterol Motil 2019; 31:e13529. [PMID: 30609268 DOI: 10.1111/nmo.13529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/24/2018] [Accepted: 11/28/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Fabry disease (FD) is a hereditary X-linked metabolic storage disorder characterized by deficient or absent lysosomal α-galactosidase A (α-Gal A) activity. This deficiency causes progressive accumulation of glycosphingolipids, primarily globotriaosylceramide (Gb3), in nearly all organ systems. Gastrointestinal (GI) symptoms can be very debilitating and are among the most frequent and earliest of the disease. As the pathophysiology of these symptoms is poorly understood, we carried out a morphological and molecular characterization of the GI tract in α-Gal A knockout mice colon in order to reveal the underlying mechanisms. METHODS Here, we performed the first morphological and biomolecular characterization of the colon wall structure in the GI tract of the α-Gal A knock-out mouse (α-Gal A -/0), a murine model of FD. KEY RESULTS Our data show a greater thickness of the gastrointestinal wall in α-Gal A (-/0) mice due to enlarged myenteric plexus' ganglia. This change is paralleled by a marked Gb3 accumulation in the gastrointestinal wall and a decreased and scattered pattern of mucosal nerve fibers. CONCLUSIONS AND INFERENCES The observed alterations are likely to be a leading cause of gut motor dysfunctions experienced by FD patients and imply that the α-Gal A (-/0) male mouse represents a reliable model for translational studies on enteropathic pain and GI symptoms in FD.
Collapse
Affiliation(s)
- Martina Masotti
- Department of Pharmacy and Biotechnology (FaBiT), Laboratory of Human and General Physiology, University of Bologna, Bologna, Italy
| | - Cecilia Delprete
- Department of Pharmacy and Biotechnology (FaBiT), Laboratory of Human and General Physiology, University of Bologna, Bologna, Italy
| | - Giovanni Dothel
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Vincenzo Donadio
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Juan Manuel Politei
- Fundation for the Study of Neurometabolic Diseases, FESEN, Buenos Aires, Argentina
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Marco Caprini
- Department of Pharmacy and Biotechnology (FaBiT), Laboratory of Human and General Physiology, University of Bologna, Bologna, Italy
| |
Collapse
|
25
|
Mazzone A, Gibbons SJ, Eisenman ST, Strege PR, Zheng T, D'Amato M, Ordog T, Fernandez-Zapico ME, Farrugia G. Direct repression of anoctamin 1 ( ANO1) gene transcription by Gli proteins. FASEB J 2019; 33:6632-6642. [PMID: 30802137 DOI: 10.1096/fj.201802373r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Ca2+-activated Cl- channel, anoctamin 1 (Ano1, also known as transmembrane protein 16A) contributes to intestinal pacemaking, fluid secretion, cellular excitability, and tissue development. The human ANO1 promoter contains binding sites for the glioma-associated oncogene (Gli) proteins. We investigated regulation of ANO1 transcription by Gli. ANO1 promoter activity was determined using a luciferase reporter system. Binding and functional effects of Glis on ANO1 transcription and expression were demonstrated by chromatin immunoprecipitation, small interfering RNA knockdown, PCR, immunolabeling, and recordings of Ca2+-activated Cl- currents in human embryonic kidney 293 (HEK293) cells. Results from previous genome-wide association studies were used to test ANO1 promoter polymorphisms for association with disease. Gli1 and Gli2 bound to the promoter and repressed ANO1 transcription. Repression depended on Gli binding to a site close to the ANO1 transcriptional start site. Mutation of this site prevented Gli binding and transcriptional repression. Knockdown of Gli expression and inhibition of Gli activity increased expression of ANO1 RNA and Ca2+-activated Cl- currents in HEK293 cells. A single-nucleotide polymorphism prevented Gli binding and showed association with irritable bowel syndrome. We conclude that Gli1 and Gli2 repress ANO1 by a novel mechanism that is independent of Gli cleavage and that has a role in gastrointestinal function.-Mazzone, A., Gibbons, S. J., Eisenman, S. T., Strege, P. R., Zheng, T., D'Amato, M., Ordog, T., Fernandez-Zapico, M. E., Farrugia, G. Direct repression of anoctamin 1 (ANO1) gene transcription by Gli proteins.
Collapse
Affiliation(s)
- Amelia Mazzone
- Enteric NeuroSciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon J Gibbons
- Enteric NeuroSciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Seth T Eisenman
- Enteric NeuroSciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter R Strege
- Enteric NeuroSciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Tenghao Zheng
- Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mauro D'Amato
- Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Biodonostia Health Research Institute, San Sebastián, Spain.,Ikerbasque-Basque Science Foundation, San Sebastián, Spain
| | - Tamas Ordog
- Enteric NeuroSciences, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
26
|
Yadak R, Breur M, Bugiani M. Gastrointestinal Dysmotility in MNGIE: from thymidine phosphorylase enzyme deficiency to altered interstitial cells of Cajal. Orphanet J Rare Dis 2019; 14:33. [PMID: 30736844 PMCID: PMC6368792 DOI: 10.1186/s13023-019-1016-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
Background MNGIE is a rare and fatal disease in which absence of the enzyme thymidine phosphorylase induces systemic accumulation of thymidine and deoxyuridine and secondary mitochondrial DNA alterations. Gastrointestinal (GI) symptoms are frequently reported in MNGIE patients, however, they are not resolved with the current treatment interventions. Recently, our understanding of the GI pathology has increased, which rationalizes the pursuit of more targeted therapeutic strategies. In particular, interstitial cells of Cajal (ICC) play key roles in GI physiology and are involved in the pathogenesis of the GI dysmotility. However, understanding of the triggers of ICC deficits in MNGIE is lacking. Herein, we review the current knowledge about the pathology of GI dysmotility in MNGIE, discuss potential mechanisms in relation to ICC loss/dysfunction, remark on the limited contribution of the current treatments, and propose intervention strategies to overcome ICC deficits. Finally, we address the advances and new research avenues offered by organoids and tissue engineering technologies, and propose schemes to implement to further our understanding of the GI pathology and utility in regenerative and personalized medicine in MNGIE. Conclusion Interstitial cells of Cajal play key roles in the physiology of the gastrointestinal motility. Evaluation of their status in the GI dysmotility related to MNGIE would be valuable for diagnosis of MNGIE. Understanding the underlying pathological and molecular mechanisms affecting ICC is an asset for the development of targeted prevention and treatment strategies for the GI dysmotility related to MNGIE.
Collapse
Affiliation(s)
- Rana Yadak
- Department of Neurology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Marjolein Breur
- Department of Child Neurology, VU University Medical center, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Abstract
Constipation, a condition characterized by heterogeneous symptoms, is common in Western society. It is associated with reduced physical health, mental health, and social functioning. Because constipation is rarely due to a life-threatening disease (for example, colon cancer), current guidelines recommend empiric therapy. Limited surveys suggest that fewer than half of treated individuals are satisfied with treatment, perhaps because the efficacy of drugs is limited, they are associated with undesirable side effects, or they may not target the underlying pathophysiology. For example, although a substantial proportion of constipated patients have a defecatory disorder that is more appropriately treated with pelvic floor biofeedback therapy than with laxatives, virtually no pharmacological trials formally assessed for anorectal dysfunction. Recent advances in investigational tools have improved our understanding of the physiology and pathophysiology of colonic and defecatory functions. In particular, colonic and anorectal high-resolution manometry are now available. High-resolution anorectal manometry, which is increasingly used in clinical practice, at least in the United States, provides a refined assessment of anorectal pressures and may uncover structural abnormalities. Advances in our understanding of colonic molecular physiology have led to the development of new therapeutic agents (such as secretagogues, pro-kinetics, inhibitors of bile acid transporters and ion exchangers). However, because clinical trials compare these newer agents with placebo, their efficacy relative to traditional laxatives is unknown. This article reviews these physiologic, diagnostic, and therapeutic advances and focuses particularly on newer therapeutic agents.
Collapse
Affiliation(s)
- David O. Prichard
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Adil E. Bharucha
- Clinical Enteric Neuroscience Translational and Epidemiological Research Program and Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| |
Collapse
|
28
|
Bonfiglio F, Henström M, Nag A, Hadizadeh F, Zheng T, Cenit MC, Tigchelaar E, Williams F, Reznichenko A, Ek WE, Rivera NV, Homuth G, Aghdassi AA, Kacprowski T, Männikkö M, Karhunen V, Bujanda L, Rafter J, Wijmenga C, Ronkainen J, Hysi P, Zhernakova A, D'Amato M. A GWAS meta-analysis from 5 population-based cohorts implicates ion channel genes in the pathogenesis of irritable bowel syndrome. Neurogastroenterol Motil 2018; 30:e13358. [PMID: 29673008 DOI: 10.1111/nmo.13358] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/23/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) shows genetic predisposition, however, large-scale, powered gene mapping studies are lacking. We sought to exploit existing genetic (genotype) and epidemiological (questionnaire) data from a series of population-based cohorts for IBS genome-wide association studies (GWAS) and their meta-analysis. METHODS Based on questionnaire data compatible with Rome III Criteria, we identified a total of 1335 IBS cases and 9768 asymptomatic individuals from 5 independent European genotyped cohorts. Individual GWAS were carried out with sex-adjusted logistic regression under an additive model, followed by meta-analysis using the inverse variance method. Functional annotation of significant results was obtained via a computational pipeline exploiting ontology and interaction networks, and tissue-specific and gene set enrichment analyses. KEY RESULTS Suggestive GWAS signals (P ≤ 5.0 × 10-6 ) were detected for 7 genomic regions, harboring 64 gene candidates to affect IBS risk via functional or expression changes. Functional annotation of this gene set convincingly (best FDR-corrected P = 3.1 × 10-10 ) highlighted regulation of ion channel activity as the most plausible pathway affecting IBS risk. CONCLUSION & INFERENCES Our results confirm the feasibility of population-based studies for gene-discovery efforts in IBS, identify risk genes and loci to be prioritized in independent follow-ups, and pinpoint ion channels as important players and potential therapeutic targets warranting further investigation.
Collapse
Affiliation(s)
- F Bonfiglio
- Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, Spain.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - M Henström
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - A Nag
- Department of Twin Research & Genetic Epidemiology, King's College London, London, England
| | - F Hadizadeh
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - T Zheng
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - M C Cenit
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - E Tigchelaar
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - F Williams
- Department of Twin Research & Genetic Epidemiology, King's College London, London, England
| | - A Reznichenko
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - W E Ek
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory Uppsala, Uppsala University, Uppsala, Sweden
| | - N V Rivera
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - G Homuth
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - A A Aghdassi
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - T Kacprowski
- Department of Functional Genomics, Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - M Männikkö
- Center for Life Course Health Research, University of Oulu, Oulu, Finland
| | - V Karhunen
- Center for Life Course Health Research, University of Oulu, Oulu, Finland.,Oulu University Hospital, Oulu, Finland.,Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - L Bujanda
- Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - J Rafter
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - C Wijmenga
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - J Ronkainen
- Center for Life Course Health Research, University of Oulu, Oulu, Finland.,Primary Health Care Center, Tornio, Finland
| | - P Hysi
- Department of Ophthalmology, King's College London, St Thomas' Hospital Campus, London, UK
| | - A Zhernakova
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - M D'Amato
- Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, Spain.,Unit of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.,BioCruces Health Research Institute, Bilbao, Spain.,IKERBASQUE, Basque Science Foundation, Bilbao, Spain
| |
Collapse
|
29
|
Bonfiglio F, Zheng T, Garcia-Etxebarria K, Hadizadeh F, Bujanda L, Bresso F, Agreus L, Andreasson A, Dlugosz A, Lindberg G, Schmidt PT, Karling P, Ohlsson B, Simren M, Walter S, Nardone G, Cuomo R, Usai-Satta P, Galeazzi F, Neri M, Portincasa P, Bellini M, Barbara G, Latiano A, Hübenthal M, Thijs V, Netea MG, Jonkers D, Chang L, Mayer EA, Wouters MM, Boeckxstaens G, Camilleri M, Franke A, Zhernakova A, D'Amato M. Female-Specific Association Between Variants on Chromosome 9 and Self-Reported Diagnosis of Irritable Bowel Syndrome. Gastroenterology 2018; 155:168-179. [PMID: 29626450 PMCID: PMC6035117 DOI: 10.1053/j.gastro.2018.03.064] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/28/2018] [Accepted: 03/31/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Genetic factors are believed to affect risk for irritable bowel syndrome (IBS), but there have been no sufficiently powered and adequately sized studies. To identify DNA variants associated with IBS risk, we performed a genome-wide association study (GWAS) of the large UK Biobank population-based cohort, which includes genotype and health data from 500,000 participants. METHODS We studied 7,287,191 high-quality single nucleotide polymorphisms in individuals who self-reported a doctor's diagnosis of IBS (cases; n = 9576) compared to the remainder of the cohort (controls; n = 336,499) (mean age of study subjects, 40-69 years). Genome-wide significant findings were further investigated in 2045 patients with IBS from tertiary centers and 7955 population controls from Europe and the United States, and a small general population sample from Sweden (n = 249). Functional annotation of GWAS results was carried out by integrating data from multiple biorepositories to obtain biological insights from the observed associations. RESULTS We identified a genome-wide significant association on chromosome 9q31.2 (single nucleotide polymorphism rs10512344; P = 3.57 × 10-8) in a region previously linked to age at menarche, and 13 additional loci of suggestive significance (P < 5.0×10-6). Sex-stratified analyses revealed that the variants at 9q31.2 affect risk of IBS in women only (P = 4.29 × 10-10 in UK Biobank) and also associate with constipation-predominant IBS in women (P = .015 in the tertiary cohort) and harder stools in women (P = .0012 in the population-based sample). Functional annotation of the 9q31.2 locus identified 8 candidate genes, including the elongator complex protein 1 gene (ELP1 or IKBKAP), which is mutated in patients with familial dysautonomia. CONCLUSIONS In a sufficiently powered GWAS of IBS, we associated variants at the locus 9q31.2 with risk of IBS in women. This observation may provide additional rationale for investigating the role of sex hormones and autonomic dysfunction in IBS.
Collapse
Affiliation(s)
- Ferdinando Bonfiglio
- Unit of Gastrointestinal Genetics, Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, San Sebastián, Spain; Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Tenghao Zheng
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Koldo Garcia-Etxebarria
- Unit of Gastrointestinal Genetics, Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, San Sebastián, Spain; Unit of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fatemeh Hadizadeh
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Luis Bujanda
- Unit of Gastrointestinal Genetics, Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, San Sebastián, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Universidad del País Vasco, San Sebastián, Spain
| | - Francesca Bresso
- Gastoenterology Unit, Tema inflammation and infection, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Agreus
- Division for Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna Andreasson
- Division for Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - Aldona Dlugosz
- Department of Medicine Solna, Karolinska Institutet, Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Greger Lindberg
- Department of Medicine Solna, Karolinska Institutet, Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Peter T Schmidt
- Department of Medicine Solna, Karolinska Institutet, Center for Digestive Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Pontus Karling
- Division of Medicine, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bodil Ohlsson
- Lund University, Skåne University Hospital, Department of Internal Medicine, Lund, Sweden
| | - Magnus Simren
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Susanna Walter
- Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Gerardo Nardone
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Rosario Cuomo
- Digestive Motility Diseases, Department of Clinical Medicine and Surgery, Federico II University Hospital, Naples, Italy
| | - Paolo Usai-Satta
- SC Gastroenterologia, Azienda Ospedaliera G. Brotzu, Cagliari, Italy
| | | | - Matteo Neri
- Department of Medicine and Aging Sciences and Center for Excellence on Aging, G. D'Annunzio University and Foundation, Chieti, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, Clinica Medica A. Murri, University of Bari Medical School, Bari, Italy
| | - Massimo Bellini
- Gastroenterology Unit, Department of Gastroenterology, University of Pisa, Pisa, Italy
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, St. Orsola, Malpighi Hospital, Bologna, Italy
| | - Anna Latiano
- Division of Gastroenterology, Istituto di Ricovero e Cura a Carattere Scientifico, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Matthias Hübenthal
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Vincent Thijs
- Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center of Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands; Department for Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Daisy Jonkers
- Department of Internal Medicine, Nutrition and Toxicology Research Institute Maastricht, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Lin Chang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, California
| | - Mira M Wouters
- Translational Research Center for Gastro Intestinal Disorders, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Center for Gastro Intestinal Disorders, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, and Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Mauro D'Amato
- Unit of Gastrointestinal Genetics, Department of Gastrointestinal and Liver Diseases, Biodonostia Health Research Institute, San Sebastián, Spain; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Unit of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Ikerbasque, Basque Science Foundation, Bilbao, Spain.
| |
Collapse
|
30
|
Strege PR, Mazzone A, Bernard CE, Neshatian L, Gibbons SJ, Saito YA, Tester DJ, Calvert ML, Mayer EA, Chang L, Ackerman MJ, Beyder A, Farrugia G. Irritable bowel syndrome patients have SCN5A channelopathies that lead to decreased Na V1.5 current and mechanosensitivity. Am J Physiol Gastrointest Liver Physiol 2018; 314:G494-G503. [PMID: 29167113 PMCID: PMC5966747 DOI: 10.1152/ajpgi.00016.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The SCN5A-encoded voltage-gated mechanosensitive Na+ channel NaV1.5 is expressed in human gastrointestinal smooth muscle cells and interstitial cells of Cajal. NaV1.5 contributes to smooth muscle electrical slow waves and mechanical sensitivity. In predominantly Caucasian irritable bowel syndrome (IBS) patient cohorts, 2-3% of patients have SCN5A missense mutations that alter NaV1.5 function and may contribute to IBS pathophysiology. In this study we examined a racially and ethnically diverse cohort of IBS patients for SCN5A missense mutations, compared them with IBS-negative controls, and determined the resulting NaV1.5 voltage-dependent and mechanosensitive properties. All SCN5A exons were sequenced from somatic DNA of 252 Rome III IBS patients with diverse ethnic and racial backgrounds. Missense mutations were introduced into wild-type SCN5A by site-directed mutagenesis and cotransfected with green fluorescent protein into HEK-293 cells. NaV1.5 voltage-dependent and mechanosensitive functions were studied by whole cell electrophysiology with and without shear force. Five of 252 (2.0%) IBS patients had six rare SCN5A mutations that were absent in 377 IBS-negative controls. Six of six (100%) IBS-associated NaV1.5 mutations had voltage-dependent gating abnormalities [current density reduction (R225W, R433C, R986Q, and F1293S) and altered voltage dependence (R225W, R433C, R986Q, G1037V, and F1293S)], and at least one kinetic parameter was altered in all mutations. Four of six (67%) IBS-associated SCN5A mutations (R225W, R433C, R986Q, and F1293S) resulted in altered NaV1.5 mechanosensitivity. In this racially and ethnically diverse cohort of IBS patients, we show that 2% of IBS patients harbor SCN5A mutations that are absent in IBS-negative controls and result in NaV1.5 channels with abnormal voltage-dependent and mechanosensitive function. NEW & NOTEWORTHY The voltage-gated Na+ channel NaV1.5 contributes to smooth muscle physiology and electrical slow waves. In a racially and ethnically mixed irritable bowel syndrome cohort, 2% had mutations in the NaV1.5 gene SCN5A. These mutations were absent in irritable bowel syndrome-negative controls. Most mutant NaV1.5 channels were loss of function in voltage dependence or mechanosensitivity.
Collapse
Affiliation(s)
- Peter R. Strege
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Amelia Mazzone
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Cheryl E. Bernard
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Leila Neshatian
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Yuri A. Saito
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - David J. Tester
- 2Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Melissa L. Calvert
- 2Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Emeran A. Mayer
- 3Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California Los Angeles, Los Angeles, California
| | - Lin Chang
- 3Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California Los Angeles, Los Angeles, California
| | - Michael J. Ackerman
- 2Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
31
|
Erickson A, Deiteren A, Harrington AM, Garcia‐Caraballo S, Castro J, Caldwell A, Grundy L, Brierley SM. Voltage-gated sodium channels: (Na V )igating the field to determine their contribution to visceral nociception. J Physiol 2018; 596:785-807. [PMID: 29318638 PMCID: PMC5830430 DOI: 10.1113/jp273461] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage-gated sodium (NaV ) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non-neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre-clinical and clinical evidence to reveal how the nine NaV channel family members (NaV 1.1-NaV 1.9) contribute to abdominal visceral function in normal and disease states.
Collapse
Affiliation(s)
- Andelain Erickson
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Annemie Deiteren
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Andrea M. Harrington
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Sonia Garcia‐Caraballo
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Ashlee Caldwell
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Luke Grundy
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| |
Collapse
|
32
|
Du P, Calder S, Angeli TR, Sathar S, Paskaranandavadivel N, O'Grady G, Cheng LK. Progress in Mathematical Modeling of Gastrointestinal Slow Wave Abnormalities. Front Physiol 2018; 8:1136. [PMID: 29379448 PMCID: PMC5775268 DOI: 10.3389/fphys.2017.01136] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
Gastrointestinal (GI) motility is regulated in part by electrophysiological events called slow waves, which are generated by the interstitial cells of Cajal (ICC). Slow waves propagate by a process of "entrainment," which occurs over a decreasing gradient of intrinsic frequencies in the antegrade direction across much of the GI tract. Abnormal initiation and conduction of slow waves have been demonstrated in, and linked to, a number of GI motility disorders. A range of mathematical models have been developed to study abnormal slow waves and applied to propose novel methods for non-invasive detection and therapy. This review provides a general outline of GI slow wave abnormalities and their recent classification using multi-electrode (high-resolution) mapping methods, with a particular emphasis on the spatial patterns of these abnormal activities. The recently-developed mathematical models are introduced in order of their biophysical scale from cellular to whole-organ levels. The modeling techniques, main findings from the simulations, and potential future directions arising from notable studies are discussed.
Collapse
Affiliation(s)
- Peng Du
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Stefan Calder
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Timothy R. Angeli
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Shameer Sathar
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | | | - Gregory O'Grady
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - Leo K. Cheng
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
- Department of Surgery, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
33
|
Cai YY, Li C, Yan ZX, Ma N, Li FF. Effects of SCN9A gene modification on Na+ channel and the expression of nerve growth factor in a rat model of diarrhea‑predominant irritable bowel syndrome. Mol Med Rep 2017; 17:1839-1846. [PMID: 29138838 DOI: 10.3892/mmr.2017.8061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 08/24/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to identify whether the sodium voltage-gated channel alpha subunit 9 (SCN9A) gene modification is a potential treatment for diarrhea‑predominant irritable bowel syndrome (D‑IBS), via regulating the Na+ channel and the expression of nerve growth factor (NGF). The recombinant adenovirus vector of the SCN9A gene was established, and rat colon cells were isolated for SCN9A gene modification. All subjects were divided into four groups: i) The SCN9A‑modified (D‑IBS rat model implanted with SCN9A‑modified colon cells), ii) negative control (NC; D‑IBS rat model implanted with colon cells without SCN9A gene modification), iii) blank (D‑IBS rat model without any treatment) and iv) normal (normal rats without any treatment). Western blotting and reverse transcription‑quantitative polymerase chain reaction were used to detect the protein and mRNA expression levels of SCN9A, NGF and voltage gated sodium channels (Nav)1.8 and Nav1.9 in rat colon tissues. Compared with the normal group, the rats in the SCN9A, NC and blank groups had significantly elevated mRNA and protein expression levels of NGF, SCN9A, Nav1.8 and Nav1.9. The rats in the SCN9A group demonstrated significantly increased mRNA and protein expression levels of NGF, SCN9A, Nav1.8 and Nav1.9 compared with the NC group and the blank group (all P<0.05). SCN9A gene modification can promote the expression of Nav1.8 and Nav1.9 channels, in addition to NGF which may provide a novel therapeutic basis for treating of D‑IBS.
Collapse
Affiliation(s)
- Yong-Yan Cai
- The First Department of Pediatric Medicinel, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Chen Li
- The Fifth Department of Pediatric Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Zhi-Xin Yan
- The First Department of Pediatric Medicinel, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Na Ma
- The First Department of Pediatric Medicinel, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Fang-Fang Li
- The First Department of Pediatric Medicinel, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
34
|
Polymer Nanodiscs: Discoidal Amphiphilic Block Copolymer Membranes as a New Platform for Membrane Proteins. Sci Rep 2017; 7:15227. [PMID: 29123151 PMCID: PMC5680229 DOI: 10.1038/s41598-017-15151-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/18/2017] [Indexed: 12/23/2022] Open
Abstract
Lipid nanodiscs are playing increasingly important roles in studies of the structure and function of membrane proteins. Development of lipid nanodiscs as a membrane-protein-supporting platform, or a drug targeting and delivery vehicle in general, is undermined by the fluidic and labile nature of lipid bilayers. Here, we report the discovery of polymer nanodiscs, i.e., discoidal amphiphilic block copolymer membrane patches encased within membrane scaffold proteins, as a novel two-dimensional nanomembrane that maintains the advantages of lipid nanodiscs while addressing their weaknesses. Using MsbA, a bacterial ATP-binding cassette transporter as a membrane protein prototype, we show that the protein can be reconstituted into the polymer nanodiscs in an active state. As with lipid nanodiscs, reconstitution of detergent-solubilized MsbA into the polymer nanodiscs significantly enhances its activity. In contrast to lipid nanodiscs that undergo time- and temperature-dependent structural changes, the polymer nanodiscs experience negligible structural evolution under similar environmental stresses, revealing a critically important property for the development of nanodisc-based characterization methodologies or biotechnologies. We expect that the higher mechanical and chemical stability of block copolymer membranes and their chemical versatility for adaptation will open new opportunities for applications built upon diverse membrane protein functions, or involved with drug targeting and delivery.
Collapse
|
35
|
Understanding unspecific complaints through genetics. Nat Genet 2016; 48:1450-1451. [PMID: 27898081 DOI: 10.1038/ng.3730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Unspecific and unexplained medical complaints can be frustrating to both patients and clinicians. A cause for these complex symptoms and signs may now have been identified.
Collapse
|
36
|
Fuentes IM, Christianson JA. Ion channels, ion channel receptors, and visceral hypersensitivity in irritable bowel syndrome. Neurogastroenterol Motil 2016; 28:1613-1618. [PMID: 27781369 PMCID: PMC5123675 DOI: 10.1111/nmo.12979] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022]
Abstract
Ion channels are expressed throughout the gastrointestinal system and regulate nearly every aspect of digestion, including fluid secretion and absorption, motility, and visceral sensitivity. It is therefore not surprising that in the setting of functional bowel disorders, such as irritable bowel syndrome (IBS), ion channels are often altered in terms of expression level and function and are a target of pharmacological intervention. This is particularly true of their role in driving abdominal pain through visceral hypersensitivity (VH), which is the main reason IBS patients seek medical care. In the study by Scanzi et al., in the current issue of this journal, they provide evidence that the T-type voltage-gated calcium channel (Cav ) Cav 3.2 is upregulated in human IBS patients, and is necessary for the induction of an IBS-like disease state in mice. In this mini-review, we will discuss the contribution of specific ion channels to VH in IBS, both in human patients and rodent models. We will also discuss how Cav 3.2 may play a role as an integrator of multiple environmental stimuli contributing toward VH.
Collapse
Affiliation(s)
- I. M. Fuentes
- Department of Anatomy and Cell Biology; School of Medicine; University of Kansas Medical Center; Kansas City KS USA
| | - J. A. Christianson
- Department of Anatomy and Cell Biology; School of Medicine; University of Kansas Medical Center; Kansas City KS USA
| |
Collapse
|
37
|
|