1
|
Aikawa G, Hoshino T, Sakuramoto H, Ouchi A, Ikeda M, Kotani M, Okamoto S, Enomoto Y, Shimojo N, Inoue Y. Quantitative visualization of gastrointestinal motility in critically ill patients using a non-invasive single-channel electro amplifier: A prospective observational cohort feasibility study. J Crit Care 2025; 87:155031. [PMID: 39893878 DOI: 10.1016/j.jcrc.2025.155031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND This study aimed to evaluate the feasibility of using electrogastrography (EGG)/electroenterography (EEnG) to quantitatively visualize gastrointestinal (GI) motor function in critically ill patients. METHODS EGG/EEnG were performed at baseline and before and after nutrition in critically ill patients with mechanical ventilation. Enteral nutrition varied in content. Dominant frequency (DF), dominant power (DP), and power ratio (PR) were calculated and compared with those from healthy controls (previous study; n = 50). RESULTS Data from 20 % of patients were unstable and could not be analyzed. Of the 54 patients analyzed, 41 were on enteral nutrition, and their age and body mass index differed from controls. Gastric DF differed significantly between critically ill patients and controls (p < 0.001). No significant difference was noted in gastric log10 DP between pre- and post-prandial periods in critically ill patients (2.79 vs 2.86, p = 0.328), but controls showed a significant increase (3.04 vs 3.22, p = 0.009). Critically ill patients had lower gastric log10 DP than controls (pre-prandial p = 0.038; post-prandial p = 0.003). In the small intestine, log10 DP did not differ significantly between pre- and post-prandial periods in critically ill patients (1.45 vs 1.52, p = 0.181), but controls showed a significant increase (1.70 vs 1.86, p < 0.001). Critically ill patients had lower small intestinal log10 DP than controls (pre-prandial p = 0.004; post-prandial p < 0.001). PR was inferior in critically ill patients than in controls. CONCLUSIONS EGG/EEnG could enable quantitative visualization of GI motor function in critically ill patients. Larger studies can determine the association of GI symptoms with risk factors and prognostic factors.
Collapse
Affiliation(s)
- Gen Aikawa
- College of Nursing, Kanto Gakuin University, Yokohama, Kanagawa, Japan; Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | - Tetsuya Hoshino
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Intensive Care, Saiseikai Yokohamashi Tobu Hospital, Yokohama, Kanagawa, Japan
| | - Hideaki Sakuramoto
- Department of Critical Care and Disaster Nursing, Japanese Red Cross Kyushu International College of Nursing, Munakata, Fukuoka, Japan
| | - Akira Ouchi
- Department of Adult Health Nursing, College of Nursing, Ibaraki Christian University, Hitachi, Ibaraki, Japan
| | - Mitsuki Ikeda
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan; Department of Nursing, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Misaki Kotani
- Department of Nursing, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Saiko Okamoto
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuki Enomoto
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Nobutake Shimojo
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiaki Inoue
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
2
|
Chunikhin AY, Danylovych HV, Danylovych YV. CHTW(R)-system in Ca 2+ transport modelling. Biosystems 2025:105456. [PMID: 40139341 DOI: 10.1016/j.biosystems.2025.105456] [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: 12/09/2024] [Revised: 03/05/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
In (Chunikhin, 2023; Chunikhin, 2024) the concept of CHTW-systems as a multidimensional representation of Petri nets was proposed based on the assumption of multidimensional distribution of tokens (resources) in positions (branes) and, accordingly, multidimensional representation of transitions and arcs. We consider the case when the main parameters of CHTW-system (threshold functions and rate functions) change in accordance with the values of the mark-functions (multidimensional resource) of some container branes of the same CHTW-system. The modification of the basic CHTW-system was designated as a CHTW(R) system, in which (R) means a Resource control of the system parameters. This approach was used to analyze changes in Ca2+ concentration in mitochondria.
Collapse
Affiliation(s)
- Alexander Yu Chunikhin
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine.
| | - Hanna V Danylovych
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Yurii V Danylovych
- Palladin Institute of Biochemistry, National Academy of Science of Ukraine, Kyiv, Ukraine
| |
Collapse
|
3
|
Bautista GM, Du Y, Matthews MJ, Flores AM, Kushnir NR, Sweeney NK, Nguyen NPN, Tokhtaeva E, Solorzano-Vargas RS, Lewis M, Stelzner M, He X, Dunn JCY, Martin MG. Smooth muscle cell Piezo1 depletion results in impaired contractile properties in murine small bowel. Commun Biol 2025; 8:448. [PMID: 40097724 PMCID: PMC11914552 DOI: 10.1038/s42003-025-07697-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Piezo1 is a mechanosensitive cation channel expressed in intestinal muscularis cells (IMCs), including smooth muscle cells (SMCs), interstitial cells of Cajal, and Pdgfrα+ cells, which form the SIP syncytium, crucial for GI contractility. Here, we investigate the effects of SMC-specific Piezo1 deletion on small bowel function. Piezo1 depletion results in weight loss, delayed GI transit, muscularis thinning, and decreased SMCs. Ex vivo analyses demonstrated impaired contractile strength and tone, while in vitro studies using IMC co-cultures show dysrhythmic Ca2+ flux with decreased frequency. Imaging reveal that Piezo1 localizes intracellularly, thereby likely impacting Ca2+ signaling mechanisms modulated by Ca2 + -handling channels located on the sarcoplasmic reticulum and plasma membrane. Our findings suggest that Piezo1 in small bowel SMCs contributes to contractility by maintaining intracellular Ca2+ activity and subsequent signaling within the SIP syncytium. These findings provide new insights into the complex role of Piezo1 in small bowel SMCs and its implications for GI motility.
Collapse
Affiliation(s)
- Geoanna M Bautista
- Department of Pediatrics, Division of Neonatology, University of California Davis Children's Hospital, Sacramento, CA, 95817, USA
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yingjie Du
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael J Matthews
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Allison M Flores
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nicole R Kushnir
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nicolle K Sweeney
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Nam Phuong N Nguyen
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Elmira Tokhtaeva
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - R S Solorzano-Vargas
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA
| | - Michael Lewis
- Department of Pathology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Matthias Stelzner
- Department of Surgery, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Ximin He
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - James C Y Dunn
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Martin G Martin
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Mattel Children's Hospital and the David Geffen School of Medicine, University of California Los Angeles, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Los Angeles, CA, 90095, USA.
| |
Collapse
|
4
|
Abstract
Gastroparesis is a neurogastrointestinal disorder of motility in which patients experience symptoms of nausea, vomiting, bloating, early satiety, postprandial fullness, upper abdominal discomfort or pain, and delayed gastric emptying of solids based on scintigraphy or stable isotope breath test when mechanical obstruction has been excluded. Symptoms of gastroparesis may result from diverse pathophysiological mechanisms, including antroduodenal hypomotility, pylorospasm, increased gastric accommodation, and visceral hypersensitivity. The most common etiologies of gastroparesis are idiopathic, diabetic, and postsurgical, and less frequent causes are neurodegenerative disorders (Parkinson's disease), myopathies (scleroderma, amyloidosis), medication-induced (glucagon-like peptide-1 agonists and opioid agents), and paraneoplastic syndrome. This review addresses pharmacologic management of gastroparesis including prokinetic and antiemetic agents, pharmacologic agents targeting the pylorus, and effects of neuromodulators. SIGNIFICANCE STATEMENT: Gastroparesis is a neurogastrointestinal motility disorder characterized by delayed gastric emptying without mechanical obstruction with numerous upper gastrointestinal symptoms, including nausea and vomiting. The management of gastroparesis involves nutritional support, medications, and procedures. The only Food and Drug Administration-approved medication for gastroparesis is metoclopramide. This article reviews the pharmacology and efficacy of all classes of antiemetics or prokinetic effects used in gastroparesis. There is still a considerable unmet need for efficacious medications specifically for the treatment of gastroparesis, especially in refractory cases.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | - Kara J Jencks
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
5
|
Simmonds S, Huizinga JD, Taberner AJ, Du P, Angeli‐Gordon TR. Electromechanical coupling across the gastroduodenal junction. Acta Physiol (Oxf) 2025; 241:e70008. [PMID: 39976325 PMCID: PMC11841026 DOI: 10.1111/apha.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/21/2025]
Abstract
The gastroduodenal junction is uniquely capable of regulating digestive functions in the gastrointestinal system. The pyloric sphincter, which demarcates the stomach from the small intestine, acts as a mechanical and electrical barrier, isolating each organ, thus enabling independent behaviors that are critical for proper digestion. Unique electrical patterns in the stomach, pylorus, and duodenum underpin the distinct contractile patterns of these regions, and improper organization of these mechanical behaviors leads to clinical conditions such as gastroparesis and dumping syndrome. For this reason, the gastroduodenal junction should be a focal point in investigations of novel biomarkers of gastrointestinal dysfunction. This review summarizes the current knowledge of bioelectrical and mechanical characteristics of the gastroduodenal junction, as well as the relevant underlying anatomy. As there is limited documentation of physiological recordings from the gastroduodenal junction of humans, inferences are made from animal studies and from measurements taken from other regions of the gastrointestinal tract, where appropriate. We suggest hypotheses on gastroduodenal electromechanical coupling and propose further studies to support or reject these ideas. Improved physiological understanding of this region, and the advent of novel diagnostic and therapeutic tools are crucial aspects for the future of clinical gastrointestinal medicine.
Collapse
Affiliation(s)
- Sam Simmonds
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
| | - Jan D. Huizinga
- Division of Gastroenterology, Department of MedicineFarncombe Family Digestive Health Research Institute, McMaster UniversityHamiltonOntarioCanada
| | - Andrew J. Taberner
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
- Department of Engineering Science and Biomedical EngineeringUniversity of AucklandAucklandNew Zealand
| | - Peng Du
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
- Department of Engineering Science and Biomedical EngineeringUniversity of AucklandAucklandNew Zealand
| | - Timothy R. Angeli‐Gordon
- Auckland Bioengineering InstituteUniversity of AucklandAucklandNew Zealand
- Department of SurgeryUniversity of AucklandAucklandNew Zealand
- Te Manawahoukura Rangahau CentreTe Wānanga o AotearoaTe AwamutuNew Zealand
| |
Collapse
|
6
|
Camillo L, Pollastro F, Talmon M, Fresu LG. Bitter Taste Receptors 38 and 46 Regulate Intestinal Peristalsis. Int J Mol Sci 2025; 26:2092. [PMID: 40076714 PMCID: PMC11900946 DOI: 10.3390/ijms26052092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Bitter taste receptors (TAS2Rs) are expressed in extraoral tissues, exerting several functions and generating a whole-body chemosensory and protective system. TAS2Rs expression has been observed in the gastrointestinal tract, although their role is poorly understood. This study aims to investigate the role of TAS2R38 and 46 in human intestinal smooth muscle cells (HISMCs) after activation with the specific bitter ligands phenylthiocarbamide and absinthin, respectively. We found that TAS2R38 and 46 activation by phenylthiocarbamide (PTC) and absinthin, respectively, induces a rapid membrane depolarization and increase of cytosolic calcium levels due to internal storage in the IP3 pathway, resulting in an accelerated cell contraction. Overall, this study unravels, for the first time, the contractile impact of these TAS2R subtypes on intestinal smooth muscle cells, suggesting their involvement in gut peristalsis and recommending these receptors as possible targets for new therapies.
Collapse
Affiliation(s)
- Lara Camillo
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (F.P.); (M.T.)
| | - Maria Talmon
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (F.P.); (M.T.)
| | - Luigia Grazia Fresu
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
7
|
Liu L, Zhao Y, Yang W, Fan Y, Han L, Sheng J, Tian Y, Gao X. Rotenone Induces Parkinsonism with Constipation Symptoms in Mice by Disrupting the Gut Microecosystem, Inhibiting the PI3K-AKT Signaling Pathway and Gastrointestinal Motility. Int J Mol Sci 2025; 26:2079. [PMID: 40076704 PMCID: PMC11899888 DOI: 10.3390/ijms26052079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. Constipation is a prodromal symptom of PD. It is important to investigate the pathogenesis of constipation symptoms in PD. Rotenone has been successfully used to establish PD animal models. However, the specific mechanism of rotenone-induced constipation symptoms is not well understood. In this work, we found that constipation symptoms appeared earlier than motor impairment in mice gavaged with a low dose of rotenone (30 mg/kg·BW). Rotenone not only caused loss of dopaminergic neurons and accumulation of α-synuclein, but also significantly reduced serum 5-HT levels and 5-HTR4 in the striatum and colon. The mRNA expression of aquaporins, gastrointestinal motility factors (c-Kit, Cx43, smMLCK and MLC-3) in mouse colon was also significantly regulated by rotenone. In addition, both colon and brain showed rotenone-induced inflammation and barrier dysfunction; the PI3K/AKT pathway in the substantia nigra and colon was also significantly inhibited by rotenone. Importantly, the structure, composition and function of the gut microbiota were also significantly altered by rotenone. Some specific taxa were closely associated with motor and constipation symptoms, inflammation, and gut and brain barrier status in PD mice. Akkermansia, Staphylococcus and Lachnospiraceae_UCG-006 may play a role in exacerbating constipation symptoms, whereas Acinetobacter, Lactobacillus, Bifidobacterium, Solibacillus and Eubacterium_xylanophilum_groups may be beneficial in stimulating gastrointestinal peristalsis, maintaining motor function and alleviating inflammation and barrier damage in mice. In conclusion, low-dose rotenone can cause parkinsonism with constipation symptoms in mice by disrupting the intestinal microecosystem and inhibiting the PI3K-AKT pathway and gastrointestinal motility.
Collapse
Affiliation(s)
- Li Liu
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (J.S.); (Y.T.)
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.Y.); (Y.F.); (L.H.)
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
| | - Yan Zhao
- Division of Science and Technology, Yunnan Agricultural University, Kunming 650201, China;
| | - Weixing Yang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.Y.); (Y.F.); (L.H.)
| | - Yuqin Fan
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.Y.); (Y.F.); (L.H.)
| | - Lixiang Han
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.Y.); (Y.F.); (L.H.)
| | - Jun Sheng
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (J.S.); (Y.T.)
| | - Yang Tian
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (J.S.); (Y.T.)
| | - Xiaoyu Gao
- Yunnan Key Laboratory of Precision Nutrition and Personalized Food Manufacturing, Yunnan Agricultural University, Kunming 650201, China; (L.L.); (J.S.); (Y.T.)
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; (W.Y.); (Y.F.); (L.H.)
- Engineering Research Center of Development and Utilization of Food and Drug Homologous Resources, Ministry of Education, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
8
|
Zhu L, Yang X. Gut Microecological Prescription: A Novel Approach to Regulating Intestinal Micro-Ecological Balance. Int J Gen Med 2025; 18:603-626. [PMID: 39931312 PMCID: PMC11807788 DOI: 10.2147/ijgm.s504616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
The intestinal microecology is comprises intestinal microorganisms and other components constituting the entire ecosystem, presenting characteristics of stability and dynamic balance. Current research reveals intestinal microecological imbalances are related to various diseases. However, fundamental research and clinical applications have not been effectively integrated. Considering the importance and complexity of regulating the intestinal microecological balance, this study provides an overview of the high-risk factors affecting intestinal microecology and detection methods. Moreover, it proposes the definition of intestinal microecological imbalance and the definition, formulation, and outcomes of gut microecological prescription to facilitate its application in clinical practice, thus promoting clinical research on intestinal microecology and improving the quality of life of the population.
Collapse
Affiliation(s)
- Lingping Zhu
- The Affiliated Nanhua Hospital, Department of General Practice, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
- School of Public Health, Fudan University, Shanghai, 200433, People’s Republic of China
| | - Xuefeng Yang
- The Affiliated Nanhua Hospital, Department of General Practice, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| |
Collapse
|
9
|
Rappold R, Kalogeropoulos K, La Regina G, auf dem Keller U, Slack E, Vogel V. Relaxation of mucosal fibronectin fibers in late gut inflammation following neutrophil infiltration in mice. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:4. [PMID: 39917413 PMCID: PMC11794144 DOI: 10.1038/s44341-024-00006-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/13/2024] [Indexed: 02/09/2025]
Abstract
The continuously remodeled extracellular matrix (ECM) plays a pivotal role in gastrointestinal health and disease, yet its precise functions remain elusive. In this study, we employed laser capture microdissection combined with low-input proteomics to investigate ECM remodeling during Salmonella-driven inflammation. To complement this, we probed how fibronectin fiber tension is altered using a mechanosensitive peptide probe. While fibronectin fibers in healthy intestinal tissue are typically stretched, many lose their tension in intestinal smooth muscles only hours after infection, despite the absence of bacteria in that area. In contrast, within the mucosa, where Salmonella is present starting 12 h post infection, fibronectin fiber relaxation occurred exclusively during late-stage infection at 72 h and was localized to already existing clusters of infiltrated neutrophils. Using N-terminomics, we identified three new cleavage sites in fibronectin in the inflamed cecum. The unique, tissue layer-specific changes in the molecular compositions and ECM fiber tension revealed herein might trigger new therapeutic strategies to fight acute intestinal inflammation.
Collapse
Affiliation(s)
- Ronja Rappold
- Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | | | - Gianna La Regina
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Ulrich auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Emma Slack
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
- Botnar Research Center for Child Health, Basel, Switzerland
| | - Viola Vogel
- Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Botnar Research Center for Child Health, Basel, Switzerland
| |
Collapse
|
10
|
Baker SA, Karwa M, Lee JY, Riar S, Drumm BT, Sanders KM. Ca²⁺ signaling in myenteric interstitial cells of Cajal (ICC-MY) and their role as conditional pacemakers in the colon. Cell Calcium 2025; 125:102990. [PMID: 39755028 PMCID: PMC11737426 DOI: 10.1016/j.ceca.2024.102990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Interstitial cells of Cajal in the plane of the myenteric plexus (ICC-MY) serve as electrical pacemakers in the stomach and small intestine. A similar population of cells is found in the colon, but these cells do not appear to generate regular slow wave potentials, as characteristic in more proximal gut regions. Ca2+ handling mechanisms in ICC-MY of the mouse proximal colon were studied using confocal imaging of muscles from animals expressing GCaMP6f exclusively in ICC. ICC-MY displayed stochastic, localized Ca2+ transients that seldom propagated between cells. Colonic ICC express ANO1 channels, so Ca2+ transients likely couple to activation of spontaneous transient inward currents (STICs) in these cells. The Ca2+ transients were due to Ca2+ release and blocked by cyclopiazonic acid (CPA), thapsigargin and caffeine, but unaffected by tetracaine. Antagonists of L- and T-type Ca2+ channels and reduction in extracellular Ca2+ had minimal effects on Ca2+ transients. We reasoned that STICs may not activate regenerative Ca2+ waves in ICC-MY because voltage-dependent Ca2+ conductances are largely inactivated at the relatively depolarized potentials of colonic muscles. We tested the effects of hyperpolarization with pinacidil, a KATP agonist. Ca2+ waves were initiated in some ICC-MY networks when muscles were hyperpolarized, and these events were blocked by a T-type Ca2+ channel antagonist, NNC 55-0396. Ca2+ waves activated by excitatory nerve stimulation were significantly enhanced by hyperpolarization. Our data suggest that colonic ICC-MY are conditional pacemaker cells that depend upon preparative hyperpolarization, produced physiologically by inputs from enteric inhibitory neurons and necessary for regenerative pacemaker activity.
Collapse
Affiliation(s)
- Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA.
| | - Manushri Karwa
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA
| | - Ji Yeon Lee
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA
| | - Sarah Riar
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dublin Road, Dundalk, A91K584, Ireland
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA.
| |
Collapse
|
11
|
Budriesi R, Corazza I, Roncioni S, Scanferlato R, De Luca D, Marzetti C, Gotti R, Rizzardi N, Bergamini C, Micucci M, Roncarati D, Mattioli LB. Herbal Extracts Mixed with Essential Oils: A Network Approach for Gastric and Intestinal Motility Disorders. Nutrients 2024; 16:4357. [PMID: 39770978 PMCID: PMC11677010 DOI: 10.3390/nu16244357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Three herbal extracts (Asparagus racemosus Willd., Tabebuia avellanedae Lorentz, and Glycyrrhiza glabra L.) were mixed with three essential oils (Foeniculum vulgare Mill., Mentha piperita L., and Pimpinella anisum L.) to formulate a product (HEMEO) whose active compounds include saponins and steroids in Asparagus racemosus, known for their anti-inflammatory properties; glycyrrhizin and flavonoids in Glycyrrhiza glabra, which exhibit gastroprotective and antispasmodic effects; menthol in Mentha piperita, contributing with antispasmodic and antimicrobial properties; and anethole and polyphenols in Pimpinella anisum, which modulate intestinal motility and offer antimicrobial activity. OBJECTIVE HEMEO was formulated for applications in intestinal motility disorders. METHODS HEMEO was evaluated for spontaneous and induced motility effects in isolated guinea pig ileum, colon, and stomach. Ex vivo experiments were conducted using LabChart software v7.0, and the product's antibacterial action against Helicobacter pylori and its antioxidant effects were assessed through disc diffusion and FRAP assays. The presence of the volatile compounds in the formulation was confirmed by GC-MS analysis; the TPC of HEMEO, determined using the Folin-Ciocalteu method, was 9.925 ± 0.42 mg GAE/g. CONCLUSIONS HEMEO showed a phenolic content correlated with its antioxidant potential and in addition inhibited H. pylori growth and demonstrated notable antioxidant properties, suggesting its role as a supportive agent in digestive processes and in managing motility disorders.
Collapse
Affiliation(s)
- Roberta Budriesi
- Food Chemistry and Nutraceutical Laboratory, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Ivan Corazza
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy;
| | - Simone Roncioni
- Valsambro S.r.l., 40121 Bologna, Italy; (S.R.); (R.S.); (D.D.L.); (C.M.)
| | | | - Dalila De Luca
- Valsambro S.r.l., 40121 Bologna, Italy; (S.R.); (R.S.); (D.D.L.); (C.M.)
| | - Carla Marzetti
- Valsambro S.r.l., 40121 Bologna, Italy; (S.R.); (R.S.); (D.D.L.); (C.M.)
| | - Roberto Gotti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (N.R.); (C.B.)
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (N.R.); (C.B.)
| | - Matteo Micucci
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy;
| | - Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Selmi 3, 40126 Bologna, Italy;
| | - Laura Beatrice Mattioli
- Food Chemistry and Nutraceutical Laboratory, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| |
Collapse
|
12
|
Koh SD, Lee JY, Ryoo SB, Drumm BT, Kim HJ, Baker SA, Sanders KM. Integrated responses of the SIP syncytium generate a major motility pattern in the colon. J Physiol 2024; 602:6659-6682. [PMID: 39572771 PMCID: PMC11908618 DOI: 10.1113/jp287315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024] Open
Abstract
The peristaltic reflex has been a central concept in gastrointestinal motility; however, evidence was published recently suggesting that post-stimulus responses that follow inhibitory neural responses provide the main propulsive force in colonic motility. This new concept was based on experiments on proximal colon where enteric inhibitory neural inputs are mainly nitrergic. However, the nature of inhibitory neural inputs changes from proximal to distal colon where purinergic inhibitory regulation dominates. In spite of the transition from nitrergic to purinergic regulation, post-stimulus responses and propulsive contractions were both blocked by antagonists of a conductance (ANO1) exclusive to interstitial cells of Cajal (ICC). How purinergic neurotransmission, transduced by PDGFRα+ cells, can influence ANO1 in ICC is unknown. We compared neural responses in proximal and distal colon. Post-stimulus responses were blocked by inhibition of nitrergic neurotransmission in proximal colon, but P2Y1 receptor antagonists were more effective in distal colon. Ca2+ entry through voltage-dependent channels (CaV3) enhances Ca2+ release in ICC. Thus, we reasoned that hyperpolarization caused by purinergic responses in PDGFRα+ cells, which are electrically coupled to ICC, might decrease inactivation of CaV3 channels and activate Ca2+ entry into ICC via anode-break upon cessation of inhibitory responses. Post-stimulus responses in distal colon were blocked by MRS2500 (P2Y1 receptor antagonist), apamin (SK channel antagonist) and NNC55-0396 (CaV3 antagonist). These compounds also blocked propagating contractions in mid and distal colon. These data provide the first clear demonstration that integration of functions in the smooth muscle-ICC-PDGFRα+ cell (SIP) syncytium generates a major motility behaviour. KEY POINTS: Propagating propulsive contractions initiated by the enteric nervous system are a major motility behaviour in the colon. A major component of contractions, necessary for propulsive contractions, occurs at cessation of enteric inhibitory neurotransmission (post-stimulus response) and is generated by interstitial cells of Cajal (ICC), which are electrically coupled to smooth muscle cells. The nature of enteric inhibitory neurotransmission shifts from proximal colon, where it is predominantly due to nitric oxide, to distal colon, where it is predominantly due to purine neurotransmitters. Different cells transduce nitric oxide and purines in the colon. ICC transduce nitric oxide, but another type of interstitial cell, PDGFRα+ cells, transduces input from purinergic neurons. However, the post-stimulus responses in proximal and distal colon are still generated in ICC. This paper explores how integrated behaviours of ICC, PDGFRα+ cells and smooth muscle cells accomplish propulsive motility in the colon.
Collapse
Affiliation(s)
- Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Ji Yeon Lee
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University Hospital, Seoul National University, Seoul, South Korea
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Hyun Jin Kim
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University, College of Medicine: Changwon, Gyeongnam-do, South Korea
| | - Sal A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
13
|
Li Y, Zhou X, Du Y, An M, Wan S, Sun Z, Zhong Q. Hesperidin facilitating gastrointestinal motility by "Gut-brain axis" and "SCF/C-Kit signaling pathways". Poult Sci 2024; 103:104390. [PMID: 39437558 PMCID: PMC11532765 DOI: 10.1016/j.psj.2024.104390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Hesperidin shows promising results as a potential feed additive for enhancing gastrointestinal motility in animals. Gastrointestinal function plays a pivotal role in animal growth and the digestibility of dietary nutrients, with gastrointestinal motor function serving as a crucial component. However, limited research has been conducted on the application of hesperidin as a feed additive to promote gastrointestinal motility. The present study aims to assess the efficacy of Hesperidin as a feed additive in promoting gastrointestinal motility and elucidating its underlying mechanism. A total of 200 newly hatched (1-day-old) broilers with similar body weight were randomly allocated into 4 groups as follows: the control group receiving only the basal diet, and the other 3 groups supplemented with 50, 100, and 150 mg of hesperidin per kg of the basal diet, respectively. Each group consisted of 5 replicates with ten broilers per replicate, and the feeding trial lasted for a duration of 21 d. At 21 d of age, a 5% w/v Evans Blue solution in distilled water was utilized to measure intestinal transit rates (ITR). Gastric emptying (GE) was evaluated by administering a phenol red solution at a concentration of 0.05% w/v (1 mL/broiler). Fifteen broilers from each group were euthanized and immediately dissected to obtain gizzard, hypothalamus, duodenum, and jugular blood samples. Jugular blood samples were collected for brain-gut peptide content analysis, while gizzard, hypothalamus, and duodenum samples were used for immunohistochemical analysis. Real-time qPCR was performed on gizzard samples. The results demonstrated a significant improvement in the GE and ITR of broilers in all treatment groups compared to the control group (P < 0.05), particularly in the 100mg/Kg and 150mg/Kg hesperidin group. Incorporation of hesperidin into the broilers' diet significantly enhances serum levels of ghrelin, encompassing serotonin (5-HT), motilin (MTL), cholecystokinin (CCK), and Stem Cell Factor (SCF) as well as substance P (SP) in the gizzard and duodenal tissues while reducing vasoactive intestinal peptide (VIP) levels (P < 0.05). The group administered a dosage of 150mg/Kg exhibited the most pronounced effect.Immunohistochemistry analysis revealed that hesperidin supplementation up-regulated SP protein content and down-regulated VIP protein content in the hypothalamus, gizzard, and duodenum of broilers (P < 0.05), with the most pronounced effect illustrated in the 150mg/Kg hesperidin group. Furthermore, addition of hesperidin to broiler feed resulted in a significant up-regulation of protein expression and gene expression related to SCF and The protein expression of Receptor tyrosine kinase (C-Kit) was significantly upregulated in the 150mg/Kg group, while the gene expression of C-Kit was significantly upregulated in the 50 mg/Kg group (P < 0.05). In conclusion, hesperidin exhibits promising potential as a feed additive for broilers, as its dietary supplementation of hesperidin improves gastrointestinal motility through modulation of both "gut-brain axis" signaling pathways and "SCF/C-Kit signaling pathways" within broiler chicken's digestive system. Notably, basal diet supplemented with 150mg/Kg hesperidin demonstrates superior efficacy.
Collapse
Affiliation(s)
- Yunfei Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xinying Zhou
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yusong Du
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Mingyuan An
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Shasha Wan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Zewei Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun 130118, China; Jilin Key Laboratory of Animal Nutrition and Feed Sciene, Jilin Agricultural University, Changchun 130118, China.
| | - Qingzhen Zhong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Changchun 130118, China; Jilin Key Laboratory of Animal Nutrition and Feed Sciene, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
14
|
Burns AJ, Goldstein AM. Causes and consequences: development and pathophysiology of Hirschsprung disease. WORLD JOURNAL OF PEDIATRIC SURGERY 2024; 7:e000903. [PMID: 39600627 PMCID: PMC11590806 DOI: 10.1136/wjps-2024-000903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Hirschsprung disease (HSCR) is a congenital enteric neuropathy in which the enteric nervous system (ENS) fails to develop along variable lengths of the distal gastrointestinal (GI) tract. This aganglionosis results in a functional bowel obstruction and requires surgical resection of the aganglionic segment. Despite surgery, however, long-term bowel dysfunction affects many patients. Understanding the embryologic causes and pathophysiologic consequences of HSCR is critical to improving its diagnosis and treatment. During normal gut development, the ENS arises from neural crest cells (NCCs) that delaminate from the neural tube to populate the entire GI tract with enteric neurons and glia. This process requires NCCs to undergo proliferation, migration and differentiation to form the complex neuroglial network that regulates gut motility and other intestinal functions. This review discusses the cellular and molecular processes that control normal ENS formation and what goes awry to give rise to HSCR. The complex pathophysiologic consequences of aganglionosis are discussed, including recent observations that describe novel aspects of HSCR beyond the absence of ganglion cells. This review aims to expand the understanding of HSCR and to stimulate new ideas on how to improve current management of the disease.
Collapse
Affiliation(s)
- Alan J Burns
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Liu D, Mao M, Liu W, Xie L, Zhong X, Cao W, Chen L. The Role of the TRPV4 Channel in Intestinal Physiology and Pathology. J Inflamm Res 2024; 17:9307-9317. [PMID: 39588136 PMCID: PMC11587805 DOI: 10.2147/jir.s483350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024] Open
Abstract
The transient receptor potential vanilloid 4 channel (TRPV4) is an important member of the TRP superfamily of cation channels. The channel can be activated by different physical and chemical stimuli, such as heat, osmotic, and mechanical stress. It regulates the release of nociceptive peptides (substance P and calcitonin gene-related peptide), and mediates neurogenic inflammation, which indicates the involvement of TRPV4 as a nociceptor. Previous studies show that TRPV4 regulates the contraction of intestinal smooth muscle, mucosal barrier permeability, intestinal ion transport, activation of submucosal enteric neurons, and generation of immune cells. TRPV4 is involved in various pathophysiological activities, and altered TRPV4 expression has been detected in some intestinal diseases (IBD, IBS, intestinal tumors, etc). Evidence indicates that TRPV4 plays a noxious role in intestinal barrier function when the intestine is damaged. This review focuses on the role of the TRPV4 channel in the physiological and pathological functions of the intestine, and evaluates the potential clinical significance to target TRPV4 channel in the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Dandan Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Mingli Mao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Wenjia Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Lihua Xie
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Xiaolin Zhong
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| | - Ling Chen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People’s Republic of China
| |
Collapse
|
16
|
Brinsi C, Jedidi S, Sammari H, Selmi H, Sebai H. Antidiarrheal, anti-inflammatory and antioxidant effects of Anethum graveolens L. fruit extract on castor oil-induced diarrhea in rats. Neurogastroenterol Motil 2024; 36:e14892. [PMID: 39115258 DOI: 10.1111/nmo.14892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/04/2024] [Accepted: 07/29/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Between food and medicine, nutraceuticals are widely used in human health for the prevention and treatment of various diseases. This study aims to determine the cytoprotective effects of Anethum gravelons fruit extract (AGFAE) on castor oil-induced diarrhea in rats due to its phytochemical and antioxidant properties. METHODS Male rats were divided into six groups of six animals each: Control (C), Castor oil (CO), CO + different doses of AGFAE (50, 100, and 200 mg/kg, b.w., p.o.), and the CO + loperamide group (LOP, 10 mg/kg, b.w., p.o.). KEY RESULTS In vitro, the chemical composition of aqueous Dill fruit extract showed strong antioxidant activity, with a high content of total polyphenols, flavonoids, and tannins. In our in vivo studies, pre-treatment with AGFAE reduced malondialdehyde and hydrogen peroxide levels and maintained normal activity of enzymatic and non-enzymatic antioxidants in the gastric and intestinal mucosa. In addition, we found that AGFAE prophylaxis improved the stability of many plasma biochemical parameters altered by castor oil intoxication, such as C-reactive protein concentrations and alkaline phosphatase activities. CONCLUSIONS & INFERENCES We suggest that AGFAE phenolic compounds had significant protection against diarrhea involving several mechanisms such as reducing hypersecretion, peristaltic, inflammation, and preserving the endogenous antioxidant levels.
Collapse
Affiliation(s)
- Chirine Brinsi
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Saber Jedidi
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
- Laboratory of Sylvo-Pastoral Resources, Institution of Agricultural Research and Higher Education (IRESA), University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka, Tunisia
| | - Houcem Sammari
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| | - Houcine Selmi
- Laboratory of Sylvo-Pastoral Resources, Institution of Agricultural Research and Higher Education (IRESA), University of Jendouba, Sylvo- Pastoral Institute of Tabarka, Tabarka, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources (LR23ES08), University of Jendouba, Higher Institute of Biotechnology of Béja, Béja, Tunisia
| |
Collapse
|
17
|
Lee J, Ko SJ, Choi NR, Choi WG, Seo M, Koo SH, Jung D, Lee MJ, Lee JH, Park JW, Kim BJ. Zuojin Pill enhances gastrointestinal motility by modulating the pacemaker potentials in interstitial cells of Cajal through multiple signaling pathways. Int J Med Sci 2024; 21:2883-2896. [PMID: 39628697 PMCID: PMC11610331 DOI: 10.7150/ijms.103507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/21/2024] [Indexed: 12/06/2024] Open
Abstract
Zuojin Pill (ZJP) is a traditional herbal preparation used to treat various gastrointestinal (GI) disorders. The purpose of this study was to elucidate the underlying cellular and molecular mechanisms by evaluating the effects of ZJP on the pacemaker activity of isolated interstitial cells of Cajal (ICCs) and on in vivo GI motility in mice. We isolated ICCs from mouse small intestine and measured pacemaker potentials by whole-cell patch clamping as well as intracellular calcium signaling by microfluorometry. Intestinal transit rate (ITR) was measured by Evans Blue migration. Administration of ZJP depolarized ICCs and reduced the amplitude and frequency of pacemaker potentials. Pretreatment with the 5-HT4 receptor antagonist RS39604, administration of the muscarinic M3 receptor antagonist 4-DAMP, or intracellular perfusion of the G-protein inhibitor GDP-β-S blocked ZJP-induced ICCs depolarization. In addition, external calcium-free medium and administration of the Ca2+-ATPase inhibitor thapsigargin, which depletes intracellular calcium stores, also blocked ZJP-induced ICCs depolarization. Moreover, ZJP-induced ICCs depolarization was inhibited in the presence of the phospholipase C (PLC) inhibitor U-73122, IP3-dependent Ca2+ release inhibitor xestospongin C, or various mitogen-activated protein kinase (MAPK) inhibitors. Alternatively, ZJP-induced ICCs depolarization in the presence of protein kinase C (PKC) inhibitors. Furthermore, ZJP reversed the reduction of ITR caused by loperamide (Imodium) and normalized the ITR abnormality of two etiologically distinct GI motility disorder (GMD) mouse models. Finally, ZJP increased serum concentrations of the pro-peristalsis factors motilin and substance P. Our results suggest that ZJP depolarizes ICCs via 5-HT4 or muscarinic M3 receptor activation and G-protein dependent calcium-, PLC-, inositol triphosphate-, and MAPK signaling pathways (but not PKC-dependent pathways), leading to enhanced GI motility.
Collapse
Affiliation(s)
- Jueun Lee
- Department of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seok Jae Ko
- Department of Clinical Korean Medicine, Graduate School of Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Na Ri Choi
- Department of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Korean Medical Science, Pusan National University School of Korean Medicine, Yangsan, Republic of Korea
| | - Woo-Gyun Choi
- Department of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Mujin Seo
- Department of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seung Hyeon Koo
- Department of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Daehwa Jung
- Department of Pharmaceutical Engineering, Daegu Hanny University, Gyeongsan 38610, Republic of Korea
| | - Min Jae Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jong Hwan Lee
- Department of Biomedical Engineering, Dong-Eui University College of Engineering, Busan 47340, Republic of Korea
| | - Jae-Woo Park
- Department of Clinical Korean Medicine, Graduate School of Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Byung Joo Kim
- Department of Longevity and Biofunctional Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
18
|
Yaguchi J, Sakai K, Horiuchi A, Yamamoto T, Yamashita T, Yaguchi S. Light-modulated neural control of sphincter regulation in the evolution of through-gut. Nat Commun 2024; 15:8881. [PMID: 39424783 PMCID: PMC11489725 DOI: 10.1038/s41467-024-53203-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024] Open
Abstract
The development of a continuous digestive tract, or through-gut, represents a key milestone in bilaterian evolution. However, the regulatory mechanisms in ancient bilaterians (urbilaterians) are not well understood. Our study, using larval sea urchins as a model, reveals a sophisticated system that prevents the simultaneous opening of the pylorus and anus, entry and exit points of the gut. This regulation is influenced by external light, with blue light affecting the pylorus via serotonergic neurons and both blue and longer wavelengths controlling the anus through cholinergic and dopaminergic neurons. These findings provide new insights into the neural orchestration of sphincter control in a simplified through-gut, which includes the esophagus, stomach, and intestine. Here, we propose that the emergence of the earliest urbilaterian through-gut was accompanied by the evolution of neural systems regulating sphincters in response to light, shedding light on the functional regulation of primordial digestive systems.
Collapse
Affiliation(s)
- Junko Yaguchi
- Shimoda Marine Research Center, University of Tsukuba, 5-10-1 Shimoda, Shizuoka, 415-0025, Japan
| | - Kazumi Sakai
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo-ku, Kyoto, Kyoto, 606-8502, Japan
| | - Atsushi Horiuchi
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo-ku, Kyoto, Kyoto, 606-8502, Japan
| | - Takashi Yamamoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| | - Takahiro Yamashita
- Department of Biophysics, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwake, Sakyo-ku, Kyoto, Kyoto, 606-8502, Japan
| | - Shunsuke Yaguchi
- Shimoda Marine Research Center, University of Tsukuba, 5-10-1 Shimoda, Shizuoka, 415-0025, Japan.
- Japan Science and Technology Agency, PRESTO, 7 Gobancho, Chiyoda-ku, 102-0076, Tokyo, Japan.
| |
Collapse
|
19
|
Frith ME, Kashyap PC, Linden DR, Theriault B, Chang EB. Microbiota-dependent early-life programming of gastrointestinal motility. iScience 2024; 27:110895. [PMID: 39351201 PMCID: PMC11440258 DOI: 10.1016/j.isci.2024.110895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/17/2023] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Gastrointestinal microbes modulate peristalsis and stimulate the enteric nervous system (ENS), whose development, as in the central nervous system (CNS), continues into the murine postweaning period. Given that adult CNS function depends on stimuli received during critical periods of postnatal development, we hypothesized that adult ENS function, namely motility, depends on microbial stimuli during similar critical periods. We gave fecal microbiota transplantation (FMT) to germ-free mice at weaning or as adults and found that only the mice given FMT at weaning recovered normal transit, while those given FMT as adults showed limited improvements. RNA sequencing (RNA-seq) of colonic muscularis propria revealed enrichments in neuron developmental pathways in mice exposed to gut microbes earlier in life, while mice exposed later-or not at all-showed exaggerated expression of inflammatory pathways. These findings highlight a microbiota-dependent sensitive period in ENS development, pointing to potential roles of the early-life microbiome in later-life dysmotility.
Collapse
Affiliation(s)
- Mary E. Frith
- Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Purna C. Kashyap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - David R. Linden
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Betty Theriault
- Department of Surgery, University of Chicago, Chicago, IL 60637, USA
| | - Eugene B. Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
20
|
Mahapatra C, Thakkar R, Kumar R. Modulatory Impact of Oxidative Stress on Action Potentials in Pathophysiological States: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1172. [PMID: 39456426 PMCID: PMC11504047 DOI: 10.3390/antiox13101172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the body's antioxidant defenses, significantly affects cellular function and viability. It plays a pivotal role in modulating membrane potentials, particularly action potentials (APs), essential for properly functioning excitable cells such as neurons, smooth muscles, pancreatic beta cells, and myocytes. The interaction between oxidative stress and AP dynamics is crucial for understanding the pathophysiology of various conditions, including neurodegenerative diseases, cardiac arrhythmias, and ischemia-reperfusion injuries. This review explores how oxidative stress influences APs, focusing on alterations in ion channel biophysics, gap junction, calcium dynamics, mitochondria, and Interstitial Cells of Cajal functions. By integrating current research, we aim to elucidate how oxidative stress contributes to disease progression and discuss potential therapeutic interventions targeting this interaction.
Collapse
Affiliation(s)
- Chitaranjan Mahapatra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ravindra Thakkar
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA
| | - Ravinder Kumar
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
21
|
Andrade ML, Herbella FAM, Patti MG, Schlottmann F. Correlation between esophageal contractility and skeletal muscle strength in healthy individuals. J Gastrointest Surg 2024; 28:1540-1542. [PMID: 38871073 DOI: 10.1016/j.gassur.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Affiliation(s)
- Marcel L Andrade
- Division of Gastroenterology, Department of Medicine, Federal University of Sergipe, Sao Cristovao, Brazil; Division of Gastrointestinal Surgery, Department of Surgery, Escola Paulista de Medicina, Federal University of São Paulo, Sao Paulo, Brazil
| | - Fernando A M Herbella
- Division of Gastrointestinal Surgery, Department of Surgery, Escola Paulista de Medicina, Federal University of São Paulo, Sao Paulo, Brazil.
| | - Marco G Patti
- Division of Gastrointestinal Surgery, Department of Surgery, Escola Paulista de Medicina, Federal University of São Paulo, Sao Paulo, Brazil
| | - Francisco Schlottmann
- Department of Surgery, University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
22
|
Danylovych HV, Danylovych YV, Pavliuk MR, Kosterin SO. Products of oxidative and non-oxidative metabolism of L-arginine as potential regulators of Ca 2+ transport in mitochondria of uterine smooth muscle. Biochim Biophys Acta Gen Subj 2024; 1868:130652. [PMID: 38857773 DOI: 10.1016/j.bbagen.2024.130652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/15/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Mitochondria play a crucial role in maintaining Ca2+ homeostasis in cells. Due to the critical regulatory role of the products of oxidative and non-oxidative metabolism of L-arginine, it is essential to clarify their effect on Ca2+ transport in smooth muscle mitochondria. Experiments were performed on the uterine myocytes of rats and isolated mitochondria. The possibility of NO synthesis by mitochondria was demonstrated by confocal microscopy and spectrofluorimetry methods using the NO-sensitive fluorescent probe DAF-FM and Mitotracker Orange CM-H2TMRos. It was shown that 50 μM L-arginine stimulates the energy-dependent accumulation of Ca2+ in mitochondria using the fluorescent probe Fluo-4 AM. A similar effect occurred when using nitric oxide donors 100 μM SNP, SNAP, and sodium nitrite (SN) directly. The stimulating effect was eliminated in the presence of the NO scavenger C-PTIO. Nitric oxide reduces the electrical potential in mitochondria without causing them to swell. The stimulatory effect of spermine on the accumulation of Ca2+ by mitochondria is attributed to the enhancement of NO synthesis, which was demonstrated with the use of C-PTIO, NO-synthase inhibitors (100 μM NA and L-NAME), as well as by direct monitoring of NO synthesis fluorescent probe DAF-FM. A conclusion was drawn about the potential regulatory effect of the product of the oxidative metabolism of L-arginine - NO on the transport of Ca2+ in the mitochondria of the myometrium, as well as the corresponding effect of the product of non-oxidative metabolism -spermine by increasing the synthesis of NO in these subcellular structures.
Collapse
Affiliation(s)
- Hanna V Danylovych
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine.
| | - Yuriy V Danylovych
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Maksym R Pavliuk
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Sergiy O Kosterin
- Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
23
|
Hwang SJ, Kim M, Jones A, Basma N, Baker SA, Sanders KM, Ward SM. Interstitial cells of the sip syncytium regulate basal membrane potential in murine gastric corpus. FASEB J 2024; 38:e23863. [PMID: 39143726 PMCID: PMC11587931 DOI: 10.1096/fj.202400982r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 08/16/2024]
Abstract
Smooth muscle cells (SMCs), Interstitial cells of Cajal (ICC) and Platelet-derived growth factor receptor α positive (PDGFRα+) cells form an integrated, electrical syncytium within the gastrointestinal (GI) muscular tissues known as the SIP syncytium. Immunohistochemical analysis of gastric corpus muscles showed that c-KIT+/ANO1+ ICC-IM and PDGFRα+ cells were closely apposed to one another in the same anatomical niches. We used intracellular microelectrode recording from corpus muscle bundles to characterize the roles of intramuscular ICC and PDGFRα+ cells in conditioning membrane potentials of gastric muscles. In muscle bundles, that have a relatively higher input impedance than larger muscle strips or sheets, we recorded an ongoing discharge of stochastic fluctuations in membrane potential, previously called unitary potentials or spontaneous transient depolarizations (STDs) and spontaneous transient hyperpolarizations (STHs). We reasoned that STDs should be blocked by antagonists of ANO1, the signature conductance of ICC. Activation of ANO1 has been shown to generate spontaneous transient inward currents (STICs), which are the basis for STDs. Ani9 reduced membrane noise and caused hyperpolarization, but this agent did not block the fluctuations in membrane potential quantitatively. Apamin, an antagonist of small conductance Ca2+-activated K+ channels (SK3), the signature conductance in PDGFRα+ cells, further reduced membrane noise and caused depolarization. Reversing the order of channel antagonists reversed the sequence of depolarization and hyperpolarization. These experiments show that the ongoing discharge of STDs and STHs by ICC and PDGFRα+ cells, respectively, exerts conditioning effects on membrane potentials in the SIP syncytium that would effectively regulate the excitability of SMCs.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| | - MinKyung Kim
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Amanda Jones
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Naseer Basma
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Salah A Baker
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Sean M Ward
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, USA
| |
Collapse
|
24
|
Shiratori R, Yokoi T, Kinoshita K, Xue W, Sasaki T, Kuga N. The Posterior Insular Cortex is Necessary for Feeding-Induced Jejunal Myoelectrical Activity in Male Rats. Neuroscience 2024; 553:40-47. [PMID: 38936460 DOI: 10.1016/j.neuroscience.2024.06.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/25/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
The gastrointestinal tract exhibits coordinated muscle motility in response to food digestion, which is regulated by the central nervous system through autonomic control. The insular cortex is one of the brain regions that may regulate the muscle motility. In this study, we examined whether, and how, the insular cortex, especially the posterior part, regulates gastrointestinal motility by recording jejunal myoelectrical signals in response to feeding in freely moving male rats. Feeding was found to induce increases in jejunal myoelectrical signal amplitudes. This increase in the jejunal myoelectrical signals was abolished by vagotomy and pharmacological inhibition of the posterior insular cortex. Additionally, feeding induced a decrease and increase in sympathetic and parasympathetic nervous activities, respectively, both of which were eliminated by posterior insular cortical inhibition. These results suggest that the posterior insular cortex regulates jejunal motility in response to feeding by modulating autonomic tone.
Collapse
Affiliation(s)
- Reina Shiratori
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Taiki Yokoi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Kosuke Kinoshita
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Wenfeng Xue
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan; Department of Neuropharmacology, Tohoku University School of Medicine, 4-1 Seiryo-machi, Aoba-Ku, Sendai 980-8575, Japan.
| | - Nahoko Kuga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan.
| |
Collapse
|
25
|
Wang Z, Liu C, Hu K, Zuo M, Tian Z, Wei Y, Zhou Q, Li Q. Postoperative delayed gastric emptying: may gut microbiota play a role? Front Cell Infect Microbiol 2024; 14:1449530. [PMID: 39193506 PMCID: PMC11347441 DOI: 10.3389/fcimb.2024.1449530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024] Open
Abstract
Postoperative delayed gastric emptying is a prevalent complication following surgical procedures, imposing heavy physical and financial burdens on patients. However, current treatment options remain suboptimal. In recent years, an increasing number of studies have highlighted that the gut microbiota and its metabolites are closely associated with postoperative complications. Various factors can disrupt the gut microbiome after surgery. This review discusses the potential mechanisms by which the gut microbiota and their metabolites may contribute to the pathogenesis of postoperative delayed gastric emptying. However, the current knowledge base is limited in terms of fully understanding the exact mechanisms involved. It is therefore evident that further research is required to fully elucidate the role of the gut microbiome in postoperative delayed gastric emptying, with the aim of uncovering new possibilities for preventive measures and therapeutic treatments.
Collapse
Affiliation(s)
- Zhiyi Wang
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
- Department of Oncology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Chuanbo Liu
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Kaiwen Hu
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Minghuan Zuo
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Zhen Tian
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Yue Wei
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Qin Zhou
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| | - Quanwang Li
- Graduate School of Beijing, University of Chinese Medicine, Beijing, China
| |
Collapse
|
26
|
Mainali BB, Yoo JJ, Ladd MR. Tissue engineering and regenerative medicine approaches in colorectal surgery. Ann Coloproctol 2024; 40:336-349. [PMID: 39228197 PMCID: PMC11375227 DOI: 10.3393/ac.2024.00437.0062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Tissue engineering and regenerative medicine (TERM) is an emerging field that has provided new therapeutic opportunities by delivering innovative solutions. The development of nontraditional therapies for previously unsolvable diseases and conditions has brought hope and excitement to countless individuals globally. Many regenerative medicine therapies have been developed and delivered to patients clinically. The technology platforms developed in regenerative medicine have been expanded to various medical areas; however, their applications in colorectal surgery remain limited. Applying TERM technologies to engineer biological tissue and organ substitutes may address the current therapeutic challenges and overcome some complications in colorectal surgery, such as inflammatory bowel diseases, short bowel syndrome, and diseases of motility and neuromuscular function. This review provides a comprehensive overview of TERM applications in colorectal surgery, highlighting the current state of the art, including preclinical and clinical studies, current challenges, and future perspectives. This article synthesizes the latest findings, providing a valuable resource for clinicians and researchers aiming to integrate TERM into colorectal surgical practice.
Collapse
Affiliation(s)
- Bigyan B Mainali
- Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Department of Biomedical Engineering, Wake Forest University, Winston-Salem, NC, USA
| | - Mitchell R Ladd
- Department of General Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
- Department of Biomedical Engineering, Wake Forest University, Winston-Salem, NC, USA
| |
Collapse
|
27
|
Wen Y, Zhan Y, Chen T, Li J, Long Q, Zheng F, Tang S, Tang X. Total Flavonoids of Aurantii Fructus Immaturus Regulate miR-5100 to Improve Constipation by Targeting Fzd2 to Alleviate Calcium Balance and Autophagy in Interstitial Cells of Cajal. Mol Neurobiol 2024; 61:5882-5900. [PMID: 38244148 DOI: 10.1007/s12035-024-03958-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Aurantii Fructus Immaturus total flavonoids (AFIF) is the main effective fraction extracted from AFI, which has a good effect on promoting gastrointestinal motility. This study aimed to investigate AFIF which regulates miR-5100 to improve constipation symptoms in mice by targeting Frizzled-2 (Fzd2) to alleviate interstitial cells of Cajal (ICCs) calcium ion balance and autophagy apoptosis. The constipated mouse model was induced by an antibiotic suspension, and then treated with AFIF. RNA-seq sequencing, luciferase assay, immunofluorescence staining, transmission electron microscopy, ELISA, flow cytometry, quantitative polymerase chain reaction (PCR), and Western blot were applied in this study. The results showed that AFIF improved constipation symptoms in antibiotic-induced constipated mice, and decreased the autophagy-related protein Beclin1 levels and the LC3-II/I ratio in ICCs. miR-5100 and its target gene Fzd2 were screened as key miRNAs and regulator associated with autophagy. Downregulation of miR-5100 caused increased expression of Fzd2, decreased proliferation activity of ICCs, increased apoptotic cells, and enhanced calcium ion release and autophagy signals. After AFIF treatment, miR-5100 expression was upregulated and Fzd2 was downregulated, while autophagy-related protein levels and calcium ion concentration decreased. Furthermore, AFIF increased the levels of SP, 5-HT, and VIP, and increased the expression of PGP9.5, Sy, and Cx43, which alleviated constipation by improving the integrity of the enteric nervous system network. In conclusion, AFIF could attenuate constipation symptoms by upregulating the expression of miR-5100 and targeting inhibition of Fzd2, alleviating calcium overload and autophagic death of ICCs, regulating the content of neurotransmitters, and enhancing the integrity of the enteric nervous system network.
Collapse
Affiliation(s)
- Yong Wen
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yu Zhan
- Anorectal Department, Affiliated Hospital of Integrative Chinese Medicine and Western Medicine of Chengdu University of TCM, Chengdu, 610042, China
- Chengdu First People's Hospital, Chengdu, 610042, China
| | - Taiyu Chen
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Nanchong, Sichuan, China
| | - Jun Li
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qing Long
- Department of Traditional Chinese Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Fan Zheng
- People's Hospital of Deyang City, Deyang, China
- Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Shiyu Tang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Xuegui Tang
- Department of Integrated Traditional and Western Medicine Anorectal, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan South Road, Nanchong, Sichuan, China.
| |
Collapse
|
28
|
Duangjai A, Rawangkan A, Yosboonruang A, Ontawong A, Saokaew S, Goh BH, Suganuma M, Phisalprapa P. Antispasmodic Activity of Light-Roasted Coffee Extract and Its Potential Use in Gastrointestinal Motility Disorders. Foods 2024; 13:2307. [PMID: 39123499 PMCID: PMC11312256 DOI: 10.3390/foods13152307] [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: 05/24/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
Antispasmodic agents are crucial in managing gastrointestinal motility disorders by modulating muscle contractions and reducing symptoms like cramping and diarrhea. This study investigated the antispasmodic potential of different coffee bean extracts, including light coffee (LC), medium coffee (MC), and dark coffee (DC), on ileum contractions induced by potassium chloride (KCl), and elucidated their mechanisms of action using in vitro isolated tissue techniques. The results demonstrated that all coffee extracts reduced spontaneous contractions of rat ileum tissue in a dose-dependent manner. Among these, LC showed the most significant reduction in ileum contractions, particularly at higher concentrations. The key findings reveal that LC at 5 mg/mL significantly reduced CaCl2-induced contractions in isolated rat ileum tissue, indicating that LC may inhibit calcium influx or interfere with calcium signaling pathways. The presence of nifedipine, propranolol, and N-nitro-L-arginine methyl ester (L-NAME) have been confirmed in their involvement; they block calcium influx and calcium channels and activate β-adrenergic pathways as part of LC's mechanism of action. The presence of their active compounds, particularly chlorogenic acid and caffeine, likely contributes to the observed antispasmodic effects. These findings suggest that LC exerts its antispasmodic effects by targeting key mechanisms involved in muscle spasms and intestinal motility, providing a potential for managing such conditions.
Collapse
Affiliation(s)
- Acharaporn Duangjai
- Unit of Excellence in Research and Product Development of Coffee, Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand;
| | - Anchalee Rawangkan
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand or (A.R.); (A.Y.)
| | - Atchariya Yosboonruang
- Division of Microbiology and Parasitology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand or (A.R.); (A.Y.)
| | - Atcharaporn Ontawong
- Unit of Excellence in Research and Product Development of Coffee, Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand;
| | - Surasak Saokaew
- Center of Health Outcomes Research and Therapeutic Safety (Cohorts), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand;
- Unit of Excellence on Clinical Outcomes Research and Integration (UNICORN), School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Unit of Excellence on Herbal Medicine, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
- Division of Pharmacy Practice, Department of Pharmaceutical Care, School of Pharmaceutical Sciences, University of Phayao, Phayao 56000, Thailand
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory Research Group (BMEX), School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia
| | - Masami Suganuma
- Graduate School of Science and Engineering, Saitama University, Saitama 3388570, Japan;
| | - Pochamana Phisalprapa
- Division of Ambulatory Medicine, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
29
|
Collier CA, Salikhova A, Sabir S, Foncerrada S, Raghavan SA. Crisis in the gut: navigating gastrointestinal challenges in Gulf War Illness with bioengineering. Mil Med Res 2024; 11:45. [PMID: 38978144 PMCID: PMC11229309 DOI: 10.1186/s40779-024-00547-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/26/2024] [Indexed: 07/10/2024] Open
Abstract
Gulf War Illness (GWI) is characterized by a wide range of symptoms that manifests largely as gastrointestinal symptoms. Among these gastrointestinal symptoms, motility disorders are highly prevalent, presenting as chronic constipation, stomach pain, indigestion, diarrhea, and other conditions that severely impact the quality of life of GWI veterans. However, despite a high prevalence of gastrointestinal impairments among these veterans, most research attention has focused on neurological disturbances. This perspective provides a comprehensive overview of current in vivo research advancements elucidating the underlying mechanisms contributing to gastrointestinal disorders in GWI. Generally, these in vivo and in vitro models propose that neuroinflammation alters gut motility and drives the gastrointestinal symptoms reported in GWI. Additionally, this perspective highlights the potential and challenges of in vitro bioengineering models, which could be a crucial contributor to understanding and treating the pathology of gastrointestinal related-GWI.
Collapse
Affiliation(s)
- Claudia A Collier
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Aelita Salikhova
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Sufiyan Sabir
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Steven Foncerrada
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
30
|
Sharma S, Buist ML. Comparing finite viscoelastic constitutive relations and variational principles in modeling gastrointestinal soft tissue deformation. J Mech Behav Biomed Mater 2024; 155:106560. [PMID: 38744120 DOI: 10.1016/j.jmbbm.2024.106560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024]
Abstract
The mechanical attributes of soft tissues within the gastrointestinal (GI) tract are crucial for the effective operation of the GI system, and alterations in these properties may play a role in motility-related disorders. Various constitutive modeling approaches have been suggested to comprehend the response of soft tissues to diverse loading conditions. Among these, hyperelastic constitutive models based on finite elasticity have gained popularity. However, these models fall short in capturing rate- and time-dependent tissue properties. In contrast, finite viscoelastic models offer a solution to overcome these limitations. Nevertheless, the development of a suitable finite viscoelastic model, coupled with a variational formulation for efficient finite element (FE) implementation, remains an ongoing challenge. This study aims to address this gap by developing diverse finite viscoelastic constitutive relations and applying them to characterize soft tissue. Furthermore, the research explores the creation of compressible, nearly incompressible, and incompressible versions of viscoelastic constitutive relations, along with their variational formulation, to facilitate efficient FE implementation. The proposed model demonstrates remarkable accuracy in replicating experimental results, achieving an R2 value exceeding 0.99.
Collapse
Affiliation(s)
- Swati Sharma
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore
| | - Martin Lindsay Buist
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore.
| |
Collapse
|
31
|
Llorente C. The Imperative for Innovative Enteric Nervous System-Intestinal Organoid Co-Culture Models: Transforming GI Disease Modeling and Treatment. Cells 2024; 13:820. [PMID: 38786042 PMCID: PMC11119846 DOI: 10.3390/cells13100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.
Collapse
Affiliation(s)
- Cristina Llorente
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
32
|
Sanketi BD, Mantri M, Huang L, Tavallaei MA, Hu S, Wang MFZ, De Vlaminck I, Kurpios NA. Villus myofibroblasts are developmental and adult progenitors of mammalian gut lymphatic musculature. Dev Cell 2024; 59:1159-1174.e5. [PMID: 38537630 PMCID: PMC11078612 DOI: 10.1016/j.devcel.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 01/26/2024] [Accepted: 03/01/2024] [Indexed: 05/09/2024]
Abstract
Inside the finger-like intestinal projections called villi, strands of smooth muscle cells contract to propel absorbed dietary fats through the adjacent lymphatic capillary, the lacteal, sending fats into the systemic blood circulation for energy production. Despite this vital function, mechanisms of formation, assembly alongside lacteals, and maintenance of villus smooth muscle are unknown. By combining single-cell RNA sequencing and quantitative lineage tracing of the mouse intestine, we identified a local hierarchy of subepithelial fibroblast progenitors that differentiate into mature smooth muscle fibers via intermediate contractile myofibroblasts. This continuum persists as the major mechanism for villus musculature renewal throughout adult life. The NOTCH3-DLL4 signaling axis governs the assembly of smooth muscle fibers alongside their adjacent lacteals and is required for fat absorption. Our studies identify the ontogeny and maintenance of a poorly defined class of intestinal smooth muscle, with implications for accelerated repair and recovery of digestive function following injury.
Collapse
Affiliation(s)
- Bhargav D Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Madhav Mantri
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Liqing Huang
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Mohammad A Tavallaei
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Michael F Z Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA
| | - Iwijn De Vlaminck
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14850, USA.
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
33
|
Kim R, Sung JH. Microfluidic gut-axis-on-a-chip models for pharmacokinetic-based disease models. BIOMICROFLUIDICS 2024; 18:031507. [PMID: 38947281 PMCID: PMC11210976 DOI: 10.1063/5.0206271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
The low success rate of new drugs transitioning from animal testing to human clinical trials necessitates the development of more accurate and representative in vitro models. Recent advances in multi-organ-on-a-chip technology offer promising avenues for studying complex organ-organ interactions. Gut-liver-on-a-chip systems hold particular promise for mimicking the intricate interplay between the gut and liver, which play crucial roles in nutrient absorption, drug metabolism, detoxification, and immune response. Here, we discuss the key components of the gut-liver axis, including the gut epithelium, liver cells, gut microbiota, and their roles in the organ functions. We then explore the potential of gut-liver-on-a-chip models to replicate the intricate interactions between the two organs for pharmacokinetic studies and their expansion to more complicated multi-organ models. Finally, we provide perspectives and future directions for developing more physiologically relevant gut-liver-axis models for more efficient drug development, studying liver diseases, and personalizing treatment strategies.
Collapse
Affiliation(s)
- Raehyun Kim
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, Republic of Korea
| |
Collapse
|
34
|
Daly AC, Cambuli F, Äijö T, Lötstedt B, Marjanovic N, Kuksenko O, Smith-Erb M, Fernandez S, Domovic D, Van Wittenberghe N, Drokhlyansky E, Griffin GK, Phatnani H, Bonneau R, Regev A, Vickovic S. Tissue and cellular spatiotemporal dynamics in colon aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590125. [PMID: 38712088 PMCID: PMC11071407 DOI: 10.1101/2024.04.22.590125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Tissue structure and molecular circuitry in the colon can be profoundly impacted by systemic age-related effects, but many of the underlying molecular cues remain unclear. Here, we built a cellular and spatial atlas of the colon across three anatomical regions and 11 age groups, encompassing ~1,500 mouse gut tissues profiled by spatial transcriptomics and ~400,000 single nucleus RNA-seq profiles. We developed a new computational framework, cSplotch, which learns a hierarchical Bayesian model of spatially resolved cellular expression associated with age, tissue region, and sex, by leveraging histological features to share information across tissue samples and data modalities. Using this model, we identified cellular and molecular gradients along the adult colonic tract and across the main crypt axis, and multicellular programs associated with aging in the large intestine. Our multi-modal framework for the investigation of cell and tissue organization can aid in the understanding of cellular roles in tissue-level pathology.
Collapse
Affiliation(s)
- Aidan C. Daly
- New York Genome Center, New York, NY, USA
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | | | - Tarmo Äijö
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Britta Lötstedt
- New York Genome Center, New York, NY, USA
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Nemanja Marjanovic
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Olena Kuksenko
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | | | | | | | | | - Eugene Drokhlyansky
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gabriel K Griffin
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hemali Phatnani
- New York Genome Center, New York, NY, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Richard Bonneau
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
- Center for Data Science, New York University, New York, NY, USA
- Current address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Aviv Regev
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Current address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Sanja Vickovic
- New York Genome Center, New York, NY, USA
- Klarman Cell Observatory Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Engineering and Herbert Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Beijer Laboratory for Gene and Neuro Research, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Chang DS, Soni KG, Preidis GA. Smooth muscle contractile responses to bile acids in mouse ileum require TGR5 but not ASBT. Front Neurol 2024; 15:1334319. [PMID: 38721114 PMCID: PMC11076673 DOI: 10.3389/fneur.2024.1334319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/08/2024] [Indexed: 05/12/2024] Open
Abstract
Background Many disorders of gut-brain interaction (DGBIs) are more prevalent in women than men and feature alterations in gastrointestinal motility and bile acid homeostasis. Mechanisms by which bile acids regulate gastrointestinal motility are poorly characterized. We recently validated an adapted tissue bath technique using everted mouse ileum, which revealed differential contractile responses to ursodeoxycholic acid (UDCA) and deoxycholic acid (DCA). Here, we aimed to determine whether these responses are dependent on host sex, the plasma membrane bile acid receptor TGR5, or the apical sodium-dependent bile acid transporter ASBT. Methods Ileal segments from male and female mice were everted and suspended in tissue baths. Contractile responses to physiologic concentrations of UDCA and DCA were quantified with or without TGR5 or ASBT inhibitors. Phosphorylation of extracellular signal-regulated kinase (ERK) and myosin light chain (MLC), markers of TGR5 activation and smooth muscle contraction, respectively, were assessed with western blot. Results There were no sex differences in the dose-dependent contractile responses to bile acids. At 100 μmol/L, UDCA but not DCA increased MLC phosphorylation and increased contractility. TGR5 inhibition decreased ERK phosphorylation and led to decreases in contractility, phosphorylated MLC, and surprisingly, total MLC. ASBT inhibition did not affect contractile responses. Conclusion Differential effects of UDCA and DCA on ileal smooth muscle contractility are not dependent on host sex or ASBT-mediated transport. Bile acids signal through mucosal TGR5, which regulates smooth muscle contractility by complex mechanisms. Understanding how bile acids differentially regulate gastrointestinal motility could facilitate new therapeutic options for specific DGBIs.
Collapse
Affiliation(s)
| | | | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
36
|
Echeverría F, Gonzalez-Sanabria N, Alvarado-Sanchez R, Fernández M, Castillo K, Latorre R. Large conductance voltage-and calcium-activated K + (BK) channel in health and disease. Front Pharmacol 2024; 15:1373507. [PMID: 38584598 PMCID: PMC10995336 DOI: 10.3389/fphar.2024.1373507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/12/2024] [Indexed: 04/09/2024] Open
Abstract
Large Conductance Voltage- and Calcium-activated K+ (BK) channels are transmembrane pore-forming proteins that regulate cell excitability and are also expressed in non-excitable cells. They play a role in regulating vascular tone, neuronal excitability, neurotransmitter release, and muscle contraction. Dysfunction of the BK channel can lead to arterial hypertension, hearing disorders, epilepsy, and ataxia. Here, we provide an overview of BK channel functioning and the implications of its abnormal functioning in various diseases. Understanding the function of BK channels is crucial for comprehending the mechanisms involved in regulating vital physiological processes, both in normal and pathological conditions, controlled by BK. This understanding may lead to the development of therapeutic interventions to address BK channelopathies.
Collapse
Affiliation(s)
- Felipe Echeverría
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Naileth Gonzalez-Sanabria
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Rosangelina Alvarado-Sanchez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Miguel Fernández
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Karen Castillo
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Investigación de Estudios Avanzados del Maule, Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
37
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
38
|
Qiao Y, Ji X, Guo H, Zheng W, Yao W. Complementary transcriptomic and proteomic analyses elucidate the toxicological molecular mechanisms of deoxynivalenol-induced contractile dysfunction in enteric smooth muscle cells. Food Chem Toxicol 2024; 186:114545. [PMID: 38403181 DOI: 10.1016/j.fct.2024.114545] [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: 01/02/2024] [Revised: 02/18/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Deoxynivalenol (DON) is one of the frequent Fusarium mycotoxins and poses a serious threat to public health worldwide. DON-induced weight loss is tightly connected with its ability to decrease feed intake by influencing gastrointestinal tract (GIT) motility. Our previous reports indicated that DON interfered with intestinal motility by injuring the contractility of enteric smooth muscle cells (SMC). Here, we further explored the potential mechanisms by employing a complementary method of transcriptomics and proteomics using the porcine enteric smooth muscle cell line (PISMC) as an experimental model. The transcriptomic and proteomic data uncover that the expression of numerous extracellular matrix (ECM) proteins and multiple integrin subunits were downregulated in PISMC under DON exposure, suppressing the ECM-integrin receptor interaction and its mediated signaling. Furthermore, DON treatment could depress actin polymerization, as reflected by the upregulated expression of Rho GTPase-activating proteins and cofilin in PISMC. Meanwhile, the expression levels of downstream contractile apparatus genes were significantly inhibited after challenge with DON. Taken together, the current results suggest that DON inhibits enteric SMC contractility by regulating the ECM-integrin-actin polymerization signaling pathway. Our findings provide novel insights into the potential mechanisms behind the DON toxicological effects in the GIT of humans and animals.
Collapse
Affiliation(s)
- Yu Qiao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
| | - Huiduo Guo
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212018, China
| | - Weijiang Zheng
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Wen Yao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
39
|
Arnette SD, Simonitis LE, Egan JP, Cohen KE, Kolmann MA. True grit? Comparative anatomy and evolution of gizzards in fishes. J Anat 2024; 244:260-273. [PMID: 37770122 PMCID: PMC10780153 DOI: 10.1111/joa.13956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
Gut morphology frequently reflects the food organisms digest. Gizzards are organs of the gut found in archosaurs and fishes that mechanically reduce food to aid digestion. Gizzards are thought to compensate for edentulism and/or provide an advantage when consuming small, tough food items (e.g., phytoplankton and algae). It is unknown how widespread gizzards are in fishes and how similar these structures are among different lineages. Here, we investigate the distribution of gizzards across bony fishes to (1) survey different fishes for gizzard presence, (2) compare the histological structure of gizzards in three species, (3) estimate how often gizzards have evolved in fishes, and (4) explore whether anatomical and ecological traits like edentulism and microphagy predict gizzard presence. According to our analyses, gizzards are rare across bony fishes, evolving only six times in a broad taxonomic sampling of 51 species, and gizzard presence is not clearly correlated with factors like gut length or dentition. We find that gizzard morphology varies among the lineages where one is present, both macroscopically (presence of a crop) and microscopically (varying tissue types). We conclude that gizzards likely aid in the mechanical reduction of food in fishes that have lost an oral dentition in their evolutionary past; however, the relative scarcity of gizzards suggests they are just one of many possible solutions for processing tough, nutrient-poor food items. Gizzards have long been present in the evolutionary history of fishes, can be found in a wide variety of marine and freshwater clades, and likely have been overlooked in many taxa.
Collapse
Affiliation(s)
- S D Arnette
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Friday Harbor Labs, University of Washington, Friday Harbor, Washington, USA
| | - L E Simonitis
- Friday Harbor Labs, University of Washington, Friday Harbor, Washington, USA
- Florida Atlantic University, Boca Raton, Florida, USA
| | - J P Egan
- Department of Biological Sciences, College of Science, University of Idaho, Moscow, Idaho, USA
- Bell Museum of Natural History, University of Minnesota, Saint Paul, Minnesota, USA
| | - K E Cohen
- University of Florida, Gainesville, Florida, USA
| | - M A Kolmann
- Department of Biology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
40
|
Idrizaj E, Nistri S, Nardini P, Baccari MC. Adiponectin affects ileal contractility of mouse preparations. Am J Physiol Gastrointest Liver Physiol 2024; 326:G187-G194. [PMID: 38111974 DOI: 10.1152/ajpgi.00203.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
Adiponectin (ADPN) has been reported to induce inhibitory effects on gastric motor activity, which, being a source of peripheral satiety signals, would contribute to the central anorexigenic effects of the hormone in rodents. However, peripheral satiety signals can also originate from the small intestine. Since there are no data on the effects of ADPN in this gut region, the present study aimed to investigate whether ADPN affects murine ileal contractility. Immunofluorescence experiments and Western blot were also performed to reveal the expression of ADPN receptors. Mechanical responses of ileal preparations were recorded in vitro via force-displacement transducers. Preparations showed a tetrodotoxin- and atropine-insensitive spontaneous contractile activity. Electrical field stimulation (EFS) induced tetrodotoxin- and atropine-sensitive contractile responses. ADPN induced a decay of the basal tension and decreased the amplitude of either the spontaneous contractility or the EFS-induced excitatory responses. All ADPN effects were abolished by the nitric oxide (NO) synthesis inhibitor NG-nitro l-arginine. The expression of the ADPN receptor, AdipoR1, but not AdipoR2, was also revealed in enteric glial cells. The present results offer the first evidence that ADPN acts on ileal preparations. The hormone exerts inhibitory effects, likely involving AdipoR1 on enteric glial cells and NO. From a physiological point of view, it could be hypothesized that the depressant action of ADPN on ileal contractility represents an additional peripheral satiety signal which, as also described for the ileal brake, could contribute to the central anorexigenic effects of the hormone.NEW & NOTEWORTHY This study provides the first evidence that adiponectin (ADPN) is able to act on ileal preparations. Functional results demonstrate that the hormone, other than causing a slight decay of the basal tension, depresses the amplitude of both spontaneous contractility and neurally induced excitatory responses of the mouse ileum through the involvement of nitric oxide. The expression of the ADPN receptor AdipoR1 and its localization on glial cells was revealed by Western blot and immunofluorescence analysis.
Collapse
Affiliation(s)
- Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Firenze, Italy
| | - Silvia Nistri
- Department of Experimental and Clinical Medicine, Imaging Platform, University of Florence, Firenze, Italy
| | - Patrizia Nardini
- Department of Experimental and Clinical Medicine, Imaging Platform, University of Florence, Firenze, Italy
| | - Maria Caterina Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Firenze, Italy
| |
Collapse
|
41
|
Vaes RDW, van Bijnen AA, Damink SWMO, Rensen SS. Pancreatic Tumor Organoid-Derived Factors from Cachectic Patients Disrupt Contractile Smooth Muscle Cells. Cancers (Basel) 2024; 16:542. [PMID: 38339292 PMCID: PMC10854749 DOI: 10.3390/cancers16030542] [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: 10/16/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
Patients with pancreatic cancer often suffer from cachexia and experience gastrointestinal symptoms that may be related to intestinal smooth muscle cell (SMC) dysfunction. We hypothesized that pancreatic tumor organoids from cachectic patients release factors that perturb the SMC's contractile characteristics. Human visceral SMCs were exposed to conditioned medium (CM) from the pancreatic tumor organoid cultures of cachectic (n = 2) and non-cachectic (n = 2) patients. Contractile proteins and markers of inflammation, muscle atrophy, and proliferation were evaluated by qPCR and Western blot. SMC proliferation and migration were monitored by live cell imaging. The Ki-67-positive cell fraction was determined in the intestinal smooth musculature of pancreatic cancer patients. CM from the pancreatic tumor organoids of cachectic patients did not affect IL-1β, IL-6, IL-8, MCP-1, or Atrogin-1 expression. However, CM reduced the α-SMA, γ-SMA, and SM22-α levels, which was accompanied by a reduced SMC doubling time and increased expression of S100A4, a Ca2+-binding protein associated with the synthetic SMC phenotype. In line with this, Ki-67-positive nuclei were increased in the intestinal smooth musculature of patients with a low versus high L3-SMI. In conclusion, patient-derived pancreatic tumor organoids release factors that compromise the contractile SMC phenotype and increase SMC proliferation. This may contribute to the frequently observed gastrointestinal motility problems in these patients.
Collapse
Affiliation(s)
- Rianne D. W. Vaes
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Annemarie A. van Bijnen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Steven W. M. Olde Damink
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of General, Visceral and Transplant Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Sander S. Rensen
- Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
42
|
Sanketi BD, Mantri M, Huang L, Tavallaei MA, Hu S, Wang MFZ, De Vlaminck I, Kurpios NA. Origin and adult renewal of the gut lacteal musculature from villus myofibroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.01.19.523242. [PMID: 36712064 PMCID: PMC9882374 DOI: 10.1101/2023.01.19.523242] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Intestinal smooth muscles are the workhorse of the digestive system. Inside the millions of finger-like intestinal projections called villi, strands of smooth muscle cells contract to propel absorbed dietary fats through the adjacent lymphatic vessel, called the lacteal, sending fats into the blood circulation for energy production. Despite this vital function, how villus smooth muscles form, how they assemble alongside lacteals, and how they repair throughout life remain unknown. Here we combine single-cell RNA sequencing of the mouse intestine with quantitative lineage tracing to reveal the mechanisms of formation and differentiation of villus smooth muscle cells. Within the highly regenerative villus, we uncover a local hierarchy of subepithelial fibroblast progenitors that progress to become mature smooth muscle fibers, via an intermediate contractile myofibroblast-like phenotype. This continuum persists in the adult intestine as the major source of renewal of villus smooth muscle cells during adult life. We further found that the NOTCH3-DLL4 signaling axis governs the assembly of villus smooth muscles alongside their adjacent lacteal, and we show that this is necessary for gut absorptive function. Overall, our data shed light on the genesis of a poorly defined class of intestinal smooth muscle and pave the way for new opportunities to accelerate recovery of digestive function by stimulating muscle repair.
Collapse
Affiliation(s)
- Bhargav D. Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Madhav Mantri
- Department of Biomedical Engineering, Cornell University; Ithaca, NY 14850, USA
| | - Liqing Huang
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Mohammad A. Tavallaei
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| | - Michael F. Z. Wang
- Department of Biomedical Engineering, Cornell University; Ithaca, NY 14850, USA
| | - Iwijn De Vlaminck
- Department of Biomedical Engineering, Cornell University; Ithaca, NY 14850, USA
| | - Natasza A. Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University; Ithaca, NY 14853, USA
| |
Collapse
|
43
|
Sanders KM, Drumm BT, Cobine CA, Baker SA. Ca 2+ dynamics in interstitial cells: foundational mechanisms for the motor patterns in the gastrointestinal tract. Physiol Rev 2024; 104:329-398. [PMID: 37561138 PMCID: PMC11281822 DOI: 10.1152/physrev.00036.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/29/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal (GI) tract displays multiple motor patterns that move nutrients and wastes through the body. Smooth muscle cells (SMCs) provide the forces necessary for GI motility, but interstitial cells, electrically coupled to SMCs, tune SMC excitability, transduce inputs from enteric motor neurons, and generate pacemaker activity that underlies major motor patterns, such as peristalsis and segmentation. The interstitial cells regulating SMCs are interstitial cells of Cajal (ICC) and PDGF receptor (PDGFR)α+ cells. Together these cells form the SIP syncytium. ICC and PDGFRα+ cells express signature Ca2+-dependent conductances: ICC express Ca2+-activated Cl- channels, encoded by Ano1, that generate inward current, and PDGFRα+ cells express Ca2+-activated K+ channels, encoded by Kcnn3, that generate outward current. The open probabilities of interstitial cell conductances are controlled by Ca2+ release from the endoplasmic reticulum. The resulting Ca2+ transients occur spontaneously in a stochastic manner. Ca2+ transients in ICC induce spontaneous transient inward currents and spontaneous transient depolarizations (STDs). Neurotransmission increases or decreases Ca2+ transients, and the resulting depolarizing or hyperpolarizing responses conduct to other cells in the SIP syncytium. In pacemaker ICC, STDs activate voltage-dependent Ca2+ influx, which initiates a cluster of Ca2+ transients and sustains activation of ANO1 channels and depolarization during slow waves. Regulation of GI motility has traditionally been described as neurogenic and myogenic. Recent advances in understanding Ca2+ handling mechanisms in interstitial cells and how these mechanisms influence motor patterns of the GI tract suggest that the term "myogenic" should be replaced by the term "SIPgenic," as this review discusses.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caroline A Cobine
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Salah A Baker
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| |
Collapse
|
44
|
Sur A, Wang Y, Capar P, Margolin G, Prochaska MK, Farrell JA. Single-cell analysis of shared signatures and transcriptional diversity during zebrafish development. Dev Cell 2023; 58:3028-3047.e12. [PMID: 37995681 PMCID: PMC11181902 DOI: 10.1016/j.devcel.2023.11.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/24/2023] [Accepted: 11/01/2023] [Indexed: 11/25/2023]
Abstract
During development, animals generate distinct cell populations with specific identities, functions, and morphologies. We mapped transcriptionally distinct populations across 489,686 cells from 62 stages during wild-type zebrafish embryogenesis and early larval development (3-120 h post-fertilization). Using these data, we identified the limited catalog of gene expression programs reused across multiple tissues and their cell-type-specific adaptations. We also determined the duration each transcriptional state is present during development and identify unexpected long-term cycling populations. Focused clustering and transcriptional trajectory analyses of non-skeletal muscle and endoderm identified transcriptional profiles and candidate transcriptional regulators of understudied cell types and subpopulations, including the pneumatic duct, individual intestinal smooth muscle layers, spatially distinct pericyte subpopulations, and recently discovered best4+ cells. To enable additional discoveries, we make this comprehensive transcriptional atlas of early zebrafish development available through our website, Daniocell.
Collapse
Affiliation(s)
- Abhinav Sur
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Yiqun Wang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Paulina Capar
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Gennady Margolin
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Morgan Kathleen Prochaska
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Jeffrey A Farrell
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA.
| |
Collapse
|
45
|
Subramanian S, Kunkel DC, Nguyen L, Coleman TP. Exploring the Gut-Brain Connection in Gastroparesis With Autonomic and Gastric Myoelectric Monitoring. IEEE Trans Biomed Eng 2023; 70:3342-3353. [PMID: 37310840 DOI: 10.1109/tbme.2023.3285491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
OBJECTIVE The goal of this study was to identify autonomic and gastric myoelectric biomarkers from throughout the day that differentiate patients with gastroparesis, diabetics without gastroparesis, and healthy controls, while providing insight into etiology. METHODS We collected 19 24-hour recordings of electrocardiogram (ECG) and electrogastrogram (EGG) data from healthy controls and patients with diabetic or idiopathic gastroparesis. We used physiologically and statistically rigorous models to extract autonomic and gastric myoelectric information from the ECG and EGG data, respectively. From these, we constructed quantitative indices which differentiated the distinct groups and demonstrated their application in automatic classification paradigms and as quantitative summary scores. RESULTS We identified several differentiators that separate healthy controls from gastroparetic patient groups, specifically around sleep and meals. We also demonstrated the downstream utility of these differentiators in automatic classification and quantitative scoring paradigms. Even with this small pilot dataset, automated classifiers achieved an accuracy of 79% separating autonomic phenotypes and 65% separating gastrointestinal phenotypes. We also achieved 89% accuracy separating controls from gastroparetic patients in general and 90% accuracy separating diabetics with and without gastroparesis. These differentiators also suggested varying etiologies for different phenotypes. CONCLUSION The differentiators we identified were able to successfully distinguish between several autonomic and gastrointestinal (GI) phenotypes using data collected while at-home with non-invasive sensors. SIGNIFICANCE Autonomic and gastric myoelectric differentiators, obtained using at-home recording of fully non-invasive signals, can be the first step towards dynamic quantitative markers to track severity, disease progression, and treatment response for combined autonomic and GI phenotypes.
Collapse
|
46
|
Sanders KM, Santana LF, Baker SA. Interstitial cells of Cajal - pacemakers of the gastrointestinal tract. J Physiol 2023. [PMID: 37997170 PMCID: PMC11908679 DOI: 10.1113/jp284745] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/06/2023] [Indexed: 11/25/2023] Open
Abstract
Gastrointestinal (GI) organs display spontaneous, non-neurogenic electrical, and mechanical rhythmicity that underlies fundamental motility patterns, such as peristalsis and segmentation. Electrical rhythmicity (aka slow waves) results from pacemaker activity generated by interstitial cells of Cajal (ICC). ICC express a unique set of ionic conductances and Ca2+ handling mechanisms that generate and actively propagate slow waves. GI smooth muscle cells lack these conductances. Slow waves propagate actively within ICC networks and conduct electrotonically to smooth muscle cells via gap junctions. Slow waves depolarize smooth muscle cells and activate voltage-dependent Ca2+ channels (predominantly CaV1.2), causing Ca2+ influx and excitation-contraction coupling. The main conductances responsible for pacemaker activity in ICC are ANO1, a Ca2+ -activated Cl- conductance, and CaV3.2. The pacemaker cycle, as currently understood, begins with spontaneous, localized Ca2+ release events in ICC that activate spontaneous transient inward currents due to activation of ANO1 channels. Depolarization activates CaV 3.2 channels, causing the upstroke depolarization phase of slow waves. The upstroke is transient and followed by a long-duration plateau phase that can last for several seconds. The plateau phase results from Ca2+ -induced Ca2+ release and a temporal cluster of localized Ca2+ transients in ICC that sustains activation of ANO1 channels and clamps membrane potential near the equilibrium potential for Cl- ions. The plateau phase ends, and repolarization occurs, when Ca2+ stores are depleted, Ca2+ release ceases and ANO1 channels deactivate. This review summarizes key mechanisms responsible for electrical rhythmicity in gastrointestinal organs.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, USA
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, USA
| | - Salah A Baker
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, USA
| |
Collapse
|
47
|
Liu G, Yu Q, Zhu H, Tan B, Yu H, Li X, Lu Y, Li H. Amyloid-β mediates intestinal dysfunction and enteric neurons loss in Alzheimer's disease transgenic mouse. Cell Mol Life Sci 2023; 80:351. [PMID: 37930455 PMCID: PMC11072809 DOI: 10.1007/s00018-023-04948-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 06/02/2023] [Accepted: 06/21/2023] [Indexed: 11/07/2023]
Abstract
Alzheimer's disease (AD) is traditionally considered as a brain disorder featured by amyloid-β (Aβ) deposition. The current study on whether pathological changes of AD extend to the enteric nervous system (ENS) is still in its infancy. In this study, we found enteric Aβ deposition, intestinal dysfunction, and colonic inflammation in the young APP/PS1 mice. Moreover, these mice exhibited cholinergic and nitrergic signaling pathways damages and enteric neuronal loss. Our data show that Aβ42 treatment remarkably affected the gene expression of cultured myenteric neurons and the spontaneous contraction of intestinal smooth muscles. The intra-colon administration of Aβ42 induced ENS dysfunction, brain gliosis, and β-amyloidosis-like changes in the wild-type mice. Our results suggest that ENS mirrors the neuropathology observed in AD brains, and intestinal pathological changes may represent the prodromal events, which contribute to brain pathology in AD. In summary, our findings provide new opportunities for AD early diagnosis and prevention.
Collapse
Affiliation(s)
- Guoqiang Liu
- Medical College, Hubei University for Nationalities, Enshi, 445000, Hubei, China
| | - Quntao Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Houze Zhu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo Tan
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyan Yu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinyan Li
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Youming Lu
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 4030030, China.
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hao Li
- Wuhan Center of Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
48
|
Rezamand G, Joukar F, Amini-Salehi E, Delam H, Zare R, Samadi A, Mavadati S, Hassanipour S, Mansour-Ghanaei F. The effectiveness of walking exercise on the bowel preparation before colonoscopy: a single blind randomized clinical trial study. BMC Gastroenterol 2023; 23:351. [PMID: 37814210 PMCID: PMC10561431 DOI: 10.1186/s12876-023-02987-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND AND AIM Bowel preparation is a crucial factor affecting the diagnostic accuracy of colonoscopy, and few randomized control trials evaluated enhancement in bowel preparation. In this study, we aimed to evaluate the effectiveness of walking exercises on bowel preparation before a colonoscopy procedure. METHODS The present study is a single-blind randomized controlled trial involving 262 patients scheduled for colonoscopy procedures. These patients were randomly assigned to two groups: an intervention group (n = 131) and a control group (n = 131). In the intervention group, participants followed a predetermined plan that included the consumption of specific liquids and foods, bisacodyl pills, polyethylene glycol powder, and a regimen of walking exercises in preparation for their colonoscopy. Conversely, individuals in the control group followed the same regimen but were not instructed to engage in walking exercises. On the day of the colonoscopy, both groups were assessed for their level of physical activity using a foot counter. Additionally, an experienced gastroenterologist evaluated and compared the bowel preparation between the two groups using the Boston Bowel Preparation Scale (BBPS). RESULTS The number of footsteps recorded in the two groups exhibited a significant difference (P < 0.001). Although there was no statistically significant difference between the intervention and control groups in terms of mean BBPS scores (6.26 ± 1.9 vs. 6.29 ± 1.9, P = 0.416), individuals who took more than 6900 steps had significantly higher BBPS scores compared to those with fewer than 6900 footsteps (6.62 ± 1.8 vs. 5.92 ± 1.9, P = 0.003).In the univariate analysis, BBPS was found to be significantly associated with individuals under the age of 50 (OR: 2.45, 95% CI: 1.30-4.61, P = 0.006) and smoking status (OR: 0.41, 95% CI: 0.17-0.94, P = 0.043). In the multivariate analysis, the relationship between BBPS and age below 50 and smoking remained significant (OR: 2.50, 95% CI: 1.30-4.70, P = 0.005, and OR: 0.38, 95% CI: 0.16-0.93, P = 0.034, respectively). CONCLUSION A higher number of footsteps taken especially more than 6900 can significantly enhance bowel preparation; however, walking exercise as an intervention before colonoscopy is not significantly associated with BBPS. Also, older people and smokers seem to have fewer benefits from walking exercises for bowel preparation. TRIAL REGISTRATION ISRCTN32724024 (Registration date:22/08/2018).
Collapse
Affiliation(s)
- Gholamreza Rezamand
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Hamed Delam
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Reza Zare
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Alireza Samadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
49
|
Story G, Briere CE, McClements DJ, Sela DA. Cannabidiol and Intestinal Motility: a Systematic Review. Curr Dev Nutr 2023; 7:101972. [PMID: 37786751 PMCID: PMC10541995 DOI: 10.1016/j.cdnut.2023.101972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/13/2023] [Accepted: 07/13/2023] [Indexed: 10/04/2023] Open
Abstract
Cannabidiol (CBD) is a non-intoxicating cannabinoid extracted from the cannabis plant that is used for medicinal purposes. Ingestion of CBD is claimed to address several pathologies, including gastrointestinal disorders, although limited evidence has been generated thus far to substantiate many of its health claims. Nevertheless, CBD usage as an over-the-counter treatment for gastrointestinal disorders is likely to expand in response to increasing commercial availability, permissive legal status, and acceptance by consumers. This systematic review critically evaluates the knowledge boundaries of the published research on CBD, intestinal motility, and intestinal motility disorders. Research on CBD and intestinal motility is currently limited but does support the safety and efficacy of CBD for several therapeutic applications, including seizure disorders, inflammatory responses, and upper gastrointestinal dysfunction (i.e., nausea and vomiting). CBD, therefore, may have therapeutic potential for addressing functional gastrointestinal disorders. The results of this review show promising in vitro and preclinical data supporting a role of CBD in intestinal motility. This includes improved gastrointestinal-related outcomes in murine models of colitis. These studies, however, vary by dose, delivery method, and CBD-extract composition. Clinical trials have yet to find a conclusive benefit of CBD on intestinal motility disorders, but these trials have been limited in scope. In addition, critical factors such as CBD dosing parameters have not yet been established. Further research will establish the efficacy of CBD in applications to address intestinal motility.
Collapse
Affiliation(s)
- Galaxie Story
- Department of Food Science, University of Massachusetts, Amherst, MA, United States
| | - Carrie-Ellen Briere
- Elaine Marieb College of Nursing, University of Massachusetts, Amherst, MA, United States
| | - D. Julian McClements
- Department of Food Science, University of Massachusetts, Amherst, MA, United States
| | - David A. Sela
- Department of Food Science, University of Massachusetts, Amherst, MA, United States
- Department of Nutrition, University of Massachusetts, Amherst, MA, United States
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
50
|
Wang Y, Jiang Y, Jiang L, Xiong W, Wang Y, Gao X, Chen Q, Lin L, Yu T, Tang Y. Estrogen increases the expression of BKCa and impairs the contraction of colon smooth muscle via upregulation of sphingosine kinase 1. J Cell Physiol 2023; 238:2390-2406. [PMID: 37642352 DOI: 10.1002/jcp.31106] [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: 06/15/2022] [Revised: 06/03/2023] [Accepted: 07/20/2023] [Indexed: 08/31/2023]
Abstract
Estrogen (E2) may impair the contraction of colonic smooth muscle (SM) leading to constipation. Large conductance Ca2+ -activated K+ channels (BKCa ) are widely expressed in the smooth muscle cells (SMCs) contributing to hyperpolarization and relaxation of SMCs. Sphingosine kinase 1 (SphK1) is known to influence the expression of BKCa . We aimed to elucidate the potential underlying molecular mechanism of BKCa and SphK1 that may influence E2-induced colonic dysmotility. In ovariectomized rats, SM contraction and expression of BKCa , SphK1, sphingosine-1-phosphate receptor (S1PR) were analyzed after the treatment with vehicle, BSA-E2, E2, and E2 receptor antagonist. The role of BKCa , SphK1, and S1PR in E2-induced SM dysmotility was investigated in rat colonic SMCs. The effect of SphK1 on SM contraction as well as on the expression of BKCa and S1PR was analyzed in SphK1 knock-out mutant mice and wild-type (WT) mice treated with or without E2. The E2-treated group exhibited a weak contraction of colonic SM and a delayed colonic transit. The treatment with E2 significantly upregulated the expression of BKCa , SphK1, S1PR1, and S1PR2, but not S1PR3, in colon SM and SMCs. Inhibition of BKCa , SphK1, S1PR1, and S1PR2 expression attenuated the effect of E2 on Ca2+ mobilization in rat colon SMCs. WT mice treated with E2 showed impaired gastrointestinal motility and enhanced expression of BKCa , S1PR1, and S1PR2 compared with those without E2 treatment. Conversely, in SphK1 knock-out mice treated with E2, these effects were partially reversed. E2 increased the release of S1P which in turn could have activated S1PR1 and S1PR2. Loss of SphK1 attenuated the effect of E2 on the upregulation of S1PR1 and S1PR2 expression. These findings indicated that E2 impaired the contraction of colon SM through activation of BKCa via the upregulation of SphK1 and the release of S1P. In the E2-induced BKCa upregulation, S1PR1 and S1PR2 might also be involved. These results may provide further insights into a therapeutic target and optional treatment approaches for patients with constipation.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ya Jiang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Jiang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjie Xiong
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanjuan Wang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangyue Gao
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qi Chen
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Lin
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Yu
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yurong Tang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|