1
|
Chen X, Song Y, Hong Y, Zhang X, Li Q, Zhou H. "NO" controversy?: A controversial role in insulin signaling of diabetic encephalopathy. Mol Cell Endocrinol 2024; 593:112346. [PMID: 39151653 DOI: 10.1016/j.mce.2024.112346] [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: 03/22/2024] [Revised: 06/14/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Insulin, a critical hormone in the human body, exerts its effects by binding to insulin receptors and regulating various cellular processes. While nitric oxide (NO) plays an important role in insulin secretion and acts as a mediator in the signal transduction pathway between upstream molecules and downstream effectors, holds a significant position in the downstream signal network of insulin. Researches have shown that the insulin-NO system exhibits a dual regulatory effect within the central nervous system, which is crucial in the regulation of diabetic encephalopathy (DE). Understanding this system holds immense practical importance in comprehending the targets of existing drugs and the development of potential therapeutic interventions. This review extensively examines the characterization of insulin, NO, Nitric oxide synthase (NOS), specific NO pathway, their interconnections, and the mechanisms underlying their regulatory effects in DE, providing a reference for new therapeutic targets of DE.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Ying Song
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China; Hangzhou King's Bio-pharmaceutical Technology Co., Ltd, Hangzhou, Zhejiang, 310007, China.
| | - Ye Hong
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xiaomin Zhang
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Qisong Li
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Hongling Zhou
- Department of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
2
|
Baccari MC, Vannucchi MG, Idrizaj E. The Possible Involvement of Glucagon-like Peptide-2 in the Regulation of Food Intake through the Gut-Brain Axis. Nutrients 2024; 16:3069. [PMID: 39339669 PMCID: PMC11435434 DOI: 10.3390/nu16183069] [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: 08/09/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Food intake regulation is a complex mechanism involving the interaction between central and peripheral structures. Among the latter, the gastrointestinal tract represents one of the main sources of both nervous and hormonal signals, which reach the central nervous system that integrates them and sends the resulting information downstream to effector organs involved in energy homeostasis. Gut hormones released by nutrient-sensing enteroendocrine cells can send signals to central structures involved in the regulation of food intake through more than one mechanism. One of these is through the modulation of gastric motor phenomena known to be a source of peripheral satiety signals. In the present review, our attention will be focused on the ability of the glucagon-like peptide 2 (GLP-2) hormone to modulate gastrointestinal motor activity and discuss how its effects could be related to peripheral satiety signals generated in the stomach and involved in the regulation of food intake through the gut-brain axis. A better understanding of the possible role of GLP-2 in regulating food intake through the gut-brain axis could represent a starting point for the development of new strategies to treat some pathological conditions, such as obesity.
Collapse
Affiliation(s)
- Maria Caterina Baccari
- Department of Experimental & Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy;
| | - Maria Giuliana Vannucchi
- Department of Experimental & Clinical Medicine, Research Unit of Histology & Embryology, University of Florence, 50139 Florence, Italy;
| | - Eglantina Idrizaj
- Department of Experimental & Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy;
| |
Collapse
|
3
|
Shaidullov I, Bouchareb D, Sorokina D, Sitdikova G. Nitric oxide in the mechanisms of inhibitory effects of sodium butyrate on colon contractions in a mouse model of irritable bowel syndrome. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03403-1. [PMID: 39191960 DOI: 10.1007/s00210-024-03403-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Irritable bowel syndrome (IBS) is a multifactorial disorder, with altered intestinal motility, visceral hypersensitivity, and dysfunction of the gut-brain axis. The aim of our study was to analyze the role of nitric oxide (NO) in the inhibitory effects of sodium butyrate on spontaneous contractility of proximal colon in a mouse model of IBS. IBS was induced by intracolonic infusion of acetic acid in the early postnatal period. Spontaneous contractions of proximal colon segments were studied in isometric conditions. The amplitude and frequency of colon contractions were higher in the IBS group. Sodium butyrate exerted inhibitory effects on colon contractions, which were less pronounced in IBS group. NO donors decreased spontaneous colon contractility and prevented the inhibitory effects of sodium butyrate in control and IBS groups. Nitric oxide synthase (NOS) inhibition by L-NAME increased contractile activity more effective in the control group and decreased the inhibitory action of sodium butyrate. In IBS group, preliminary application of L-NAME did not prevent sodium butyrate action. Our data indicate that butyrate exerts its inhibitory effects on colon motility at least partially through activation of NO synthesis. In the IBS model group, the NO-dependent mechanisms were less effective probably due to downregulation of NOS.
Collapse
Affiliation(s)
- Ilnar Shaidullov
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18, Kremlevskaya Str, 420008, Kazan, Russia.
| | - Djamila Bouchareb
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18, Kremlevskaya Str, 420008, Kazan, Russia
| | - Dina Sorokina
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18, Kremlevskaya Str, 420008, Kazan, Russia
| | - Guzel Sitdikova
- Department of Human and Animal Physiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18, Kremlevskaya Str, 420008, Kazan, Russia
| |
Collapse
|
4
|
Hung JH, Zhang SM, Huang SL. Nitrate promotes the growth and the production of short-chain fatty acids and tryptophan from commensal anaerobe Veillonella dispar in the lactate-deficient environment by facilitating the catabolism of glutamate and aspartate. Appl Environ Microbiol 2024; 90:e0114824. [PMID: 39082806 PMCID: PMC11337843 DOI: 10.1128/aem.01148-24] [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: 06/15/2024] [Accepted: 07/10/2024] [Indexed: 08/22/2024] Open
Abstract
Veillonella spp. are nitrate-reducing bacteria with anaerobic respiratory activity that reduce nitrate to nitrite. They are obligate anaerobic, Gram-negative cocci that ferment lactate as the main carbon source and produce short-chain fatty acids (SCFAs). Commensal Veillonella reside in the human body site where lactate level is, however, limited for Veillonella growth. In this study, nitrate was shown to promote the anaerobic growth of Veillonella in the lactate-deficient media. We aimed to investigate the underlying mechanisms and the metabolism involved in nitrate respiration. Nitrate (15 mM) was demonstrated to promote Veillonella dispar growth and viability in the tryptone-yeast extract medium containing 0.5 mM L-lactate. Metabolite and transcriptomic analyses revealed nitrate enabled V. dispar to actively utilize glutamate and aspartate from the medium and secrete tryptophan. Glutamate or aspartate was further supplemented to a medium to investigate individual catabolism during nitrate respiration. Notably, nitrate was demonstrated to elevate SCFA production in the glutamate-supplemented medium, and further increase tryptophan production in the aspartate-supplemented medium. We proposed that the increased consumption of glutamate provided reducing power for nitrate respiration and aspartate served as a substrate for fumarate formation. Both glutamate and aspartate were incorporated into the central metabolic pathways via reverse tricarboxylic acid cycle and were linked with the increased production of acetate, propionate, and tryptophan. This study provides further understanding of the promoted growth and metabolic mechanisms by commensal V. dispar utilizing nitrate and specific amino acids to adapt to the lactate-deficient environment.IMPORTANCENitrate is a pivotal ecological factor influencing microbial community and metabolism. Dietary nitrate provides health benefits including anti-diabetic and anti-hypertensive effects via microbial-derived metabolites such as nitrite. Unraveling the impacts of nitrate on the growth and metabolism of human commensal bacteria is imperative to comprehend the intricate roles of nitrate in regulating microbial metabolism, community, and human health. Veillonella are lactate-utilizing, nitrate-reducing bacteria that are frequently found in the human body site where lactate levels are low and nitrate is at millimolar levels. Here, we comprehensively described the metabolic strategies employed by V. dispar to thrive in the lactate-deficient environment using nitrate respiration and catabolism of specific amino acids. The elevated production of SCFAs and tryptophan from amino acids during nitrate respiration of V. dispar further suggested the potential roles of nitrate and Veillonella in the promotion of human health.
Collapse
Affiliation(s)
- Jia-He Hung
- School of Medicine, National Yang Ming Chiao Tung University, Yangming Campus, Taipei, Taiwan
| | - Shi-Min Zhang
- Program in Molecular Medicine, National Yang Ming Chiao Tung University, Yangming Campus, Taipei, Taiwan
| | - Shir-Ly Huang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Yangming Campus, Taipei, Taiwan
| |
Collapse
|
5
|
Cai T, Dong Y, Feng Z, Cai B. Ameliorative effects of the mixed aqueous extract of Aurantii Fructus Immaturus and Magnoliae Officinalis Cortex on loperamide-induced STC mice. Heliyon 2024; 10:e33705. [PMID: 39040398 PMCID: PMC11261063 DOI: 10.1016/j.heliyon.2024.e33705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
Aurantii fructus immaturus (AFI) and Magnoliae Officinalis Cortex (MOC) have been used to treat constipation in China for thousands of years. In this study, a mouse model of slow transit constipation (STC) was established by gavage of loperamide at a dose of 10 mg/kg bw/day for seven days. Seventy-two mice were randomly allocated to six groups (control, STC model, 3 g/kg AFI + MOC, 6 g/kg AFI + MOC, 12 g/kg AFI + MOC, and mosapride). A mixed aqueous extract of AFI and MOC was administered to the STC mice at the corresponding doses from the first day of modelling. Body weight, faecal water content, gastrointestinal transit time, and intestinal propulsion rate were evaluated. Serum levels of neurotransmitters and gastrointestinal hormones, colonic expression of aquaporins (AQP), and interstitial cells of Cajal (ICC) were assessed using ELISA, immunohistochemistry, and Western blot analysis. The abundance and diversity of the gut microbiota were analysed by 16S rRNA gene sequencing. The mixed aqueous extract significantly increased faecal water content and intestinal propulsion rate and shortened gastrointestinal transit time in STC mice. Furthermore, the administration of AFI and MOC significantly decreased serum vasoactive intestinal peptide (VIP), nitric oxide (NO), and somatostatin (SS) levels and increased serum motilin (MTL) levels in STC mice. The protein expression levels of AQP3 and AQP4 in the colon tissue of STC mice significantly decreased following AFI + MOC treatment, whereas those of AQP9 significantly increased. Moreover, the AFI + MOC treatment led to an increase in the number and functionality of ICCs. In addition, the relative abundances of Ruminococcus and Oscillospira increased in response to the administration of AFI + MOC in STC mice. In conclusion, the mixed aqueous extract of AFI and MOC promoted defaecation and increased intestinal mobility in STC mice. Its mechanisms of action involve modulatory effects on neurotransmitters, gastrointestinal hormones, AQPs, and ICCs. AFI + MOC treatment also improved the diversity and abundance of the gut microbiota in STC mice, particularly short-chain fatty acid-producing bacteria, which may play an important role in its beneficial effect on constipation.
Collapse
Affiliation(s)
- Ting Cai
- Department of Nephrology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Wuxi, 214000, China
| | - Yun Dong
- Department of Pathology, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Zeyu Feng
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Bin Cai
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| |
Collapse
|
6
|
Zhang C, Zhang R, Cheng Y, Chen J, Zhu R, Gao L, Han M. Role of Zhiqiao Chuanlian decoction in the treatment of food accumulation fever: Network pharmacology and animal experiments. Heliyon 2024; 10:e29813. [PMID: 38681542 PMCID: PMC11053291 DOI: 10.1016/j.heliyon.2024.e29813] [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: 10/04/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Objective Food accumulation fever (FAF), a common clinical disease in children, is generally induced by the excessive intake of high-calorie or high-fat foods. Zhiqiao Chuanlian decoction (ZQCLD) is a classical traditional Chinese medicine (TCM) that may have therapeutic effects on FAF. Methods Network pharmacological analyses of ZQCLD and FAF were conducted. Animal experiments lasted for 14 days. Rats in the model, positive control, and low-, medium-, and high-dose groups were fed a high-calorie diet. On days 11-14, the positive group was given a domperidone solution. The low-, medium-, and high-dose groups were administered different concentrations of ZQCLD. The body temperature, gastric emptying rate, and intestinal propulsion rate were measured. Relevant indicators were determined by ELISA. Results The main target proteins included IL-1β, C-C motif chemokine 2 (CCL2), prostaglandin G/H synthase 2 (PTGS2), transcription factor AP-1 (JUN), haem oxygenase 1 (HMOX1), interferon-gamma (IFN-γ), peroxisome proliferator-activated receptor-gamma (PPAR-γ), and inducible nitric oxide synthase (NOS2/iNOS). Compared with those in the control group, body weight, gastric emptying rate, intestinal propulsion rate, and neuronal nitric oxide synthase (NOS1/nNOS) levels were significantly lower in the model group, whereas body temperature and endotoxin, interleukin-1β (IL-1β), PGE2, and iNOS levels were increased. In each treatment group, body temperature and PGE2 levels returned to normal levels. Compared with those in the model group, the gastric emptying rates in the positive group and the low- and medium-dose groups increased; the intestinal propulsion rates were higher in the medium- and high-dose groups, whereas the endotoxin and IL-1β levels were lower; and the nNOS level was higher in the high-dose group, whereas the iNOS level was lower. Conclusions ZQCLD may treat FAF by regulating jejunal IL-1β and nNOS, serum endotoxin, and hypothalamic PGE2 and iNOS levels.
Collapse
Affiliation(s)
- Chuxin Zhang
- Qi-Huang Chinese Medicine School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Ruoshi Zhang
- Qi-Huang Chinese Medicine School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Yuli Cheng
- Qi-Huang Chinese Medicine School, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Jingpeng Chen
- The Second Clinical Medical College, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Ruizi Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Lin Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 100029, Beijing, China
| | - Mei Han
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, 100029, Beijing, China
| |
Collapse
|
7
|
Sun Q, Gao N, Song J, Jia J, Dong A, Xia W. The association between tea consumption and non-malignant digestive system diseases: A Mendelian randomized study. Clin Nutr ESPEN 2024; 60:327-332. [PMID: 38479931 DOI: 10.1016/j.clnesp.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/27/2024] [Accepted: 02/09/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Tea consumption might be closely related to non-malignant digestive diseases. Nevertheless, this correlation remains inadequately comprehended. Therefore, our objective was to elucidate the essence of these connections. METHODS This study employed a Mendelian randomization approach to investigate the impact of tea consumption on specific digestive disorders. Genetic data associated with tea consumption were obtained from the UK Biobank (UKB), encompassing 447,485 participants. We chose a gene-wide association study with no sample overlap and UKB as our data source for all outcomes. The primary analytical method utilized was inverse variance weighting, and multiple analytical models were employed to enhance the analysis's reliability and ensure robust results. RESULT Our investigation revealed that tea consumption was linked to an elevated susceptibility to gastroesophageal reflux disease (GERD). However, there was a lack of substantial evidence suggesting an association between tea intake and Crohn's disease (CD), ulcerative colitis (UC), or non-alcoholic fatty liver disease (NAFLD). CONCLUSIONS Our study suggests that the excessive consumption of tea may heighten the likelihood of GERD. These results hold potential significance in guiding dietary pattern modifications for individuals with GERD. Furthermore, there may be value in implementing GERD monitoring and preventive measures in populations with elevated tea consumption.
Collapse
Affiliation(s)
- Qi Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
| | - Ning Gao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangwei Song
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Junjun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China
| | - Aiqiang Dong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Weiliang Xia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, School of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou 310003, China.
| |
Collapse
|
8
|
Bhowmik R, Roy M. Recent advances on the development of NO-releasing molecules (NORMs) for biomedical applications. Eur J Med Chem 2024; 268:116217. [PMID: 38367491 DOI: 10.1016/j.ejmech.2024.116217] [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: 12/11/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Abstract
Nitric oxide (NO) is an important biological messenger as well as a signaling molecule that participates in a broad range of physiological events and therapeutic applications in biological systems. However, due to its very short half-life in physiological conditions, its therapeutic applications are restricted. Efforts have been made to develop an enormous number of NO-releasing molecules (NORMs) and motifs for NO delivery to the target tissues. These NORMs involve organic nitrate, nitrite, nitro compounds, transition metal nitrosyls, and several nanomaterials. The controlled release of NO from these NORMs to the specific site requires several external stimuli like light, sound, pH, heat, enzyme, etc. Herein, we have provided a comprehensive review of the biochemistry of nitric oxide, recent advancements in NO-releasing materials with the appropriate stimuli of NO release, and their biomedical applications in cancer and other disease control.
Collapse
Affiliation(s)
- Rintu Bhowmik
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur, India.
| |
Collapse
|
9
|
Koc A, Koc DS, Askin CI, Kara H, Ozturk Fincan GS, Ozger Ilhan S, Sarioglu Y. Effects of hydrogen sulfide on relaxation responses in the lower esophageal sphincter in rabbits: the potential role of potassium channels. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1537-1550. [PMID: 37668686 DOI: 10.1007/s00210-023-02695-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023]
Abstract
Hydrogen sulfide (H2S) is a significant physiologic inhibitory neurotransmitter. The main goal of this research was to examine the contribution of diverse potassium (K+) channels and nitric oxide (NO) in mediating the H2S effect on electrical field stimulation (EFS)-induced neurogenic contractile responses in the lower esophageal sphincter (LES). EFS-induced contractile responses of rabbit isolated LES strips were recorded using force transducers in organ baths that contain Krebs-Henseleit solutions (20 ml). Cumulative doses of NaHS, L-cysteine, PAG, and AOAA were evaluated in NO-dependent and NO-independent groups. The experiments were conducted again in the presence of K+ channel blockers. In both NO-dependent and NO-independent groups, NaHS, L-cysteine, PAG, and AOAA significantly reduced EFS-induced contractile responses. In the NO-dependent group, the effect of NaHS and L-cysteine decreased in the presence of 4-AP, and also the effect of NaHS decreased in the NO-dependent and independent group in the presence of TEA. In the NO-independent group, K+ channel blockers didn't change L-cysteine-induced relaxations. K+ channel blockers had no impact on the effects of PAG and AOAA. In addition, NaHS significantly relaxed 80-mM KCl-induced contractions, whereas L-cysteine, PAG, and AOAA did not. In the present study, H2S decreased the amplitudes of EFS-induced contraction responses. These results suggest that Kv channels and NO significantly contribute to exogenous H2S and endogenous H2S precursor L-cysteine inhibitory effect on lower esophageal sphincter smooth muscle.
Collapse
Affiliation(s)
- Aysegul Koc
- Department of Medical Pharmacology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Turkey
| | - Derya Sebile Koc
- Department of Medical Pharmacology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Celil Ilker Askin
- Department of Medical Pharmacology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Halil Kara
- Department of Medical Pharmacology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara, Turkey.
| | | | - Sevil Ozger Ilhan
- Department of Medical Pharmacology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Yusuf Sarioglu
- Department of Medical Pharmacology, Faculty of Medicine, Istinye University, Istanbul, Turkey
| |
Collapse
|
10
|
Idrizaj E, Biagioni C, Traini C, Vannucchi MG, Baccari MC. Glucagon-like Peptide-2 Depresses Ileal Contractility in Preparations from Mice through Opposite Modulatory Effects on Nitrergic and Cholinergic Neurotransmission. Int J Mol Sci 2024; 25:1855. [PMID: 38339131 PMCID: PMC10855646 DOI: 10.3390/ijms25031855] [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: 12/07/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
Glucagon-like peptide-2 (GLP-2) has been reported to influence gastrointestinal motor responses, exerting a modulatory role on enteric neurotransmission. To our knowledge, no data on GLP-2 effects on the motility of the isolated ileum are available; therefore, we investigated whether GLP-2 affects the contractile activity of mouse ileal preparations and the neurotransmitters engaged. Ileal preparations showed tetrodotoxin (TTX)- and atropine-insensitive spontaneous contractile activity, which was unaffected by the nitric oxide synthesis inhibitor, L-NNA. GLP-2 depressed the spontaneous contractility, an effect that was abolished by TTX or L-NNA and not influenced by atropine. Electrical field stimulation induced TTX- and atropine-sensitive contractile responses, which were reduced in amplitude by GLP-2 even in the presence of L-NNA. Immunohistochemical results showed a significant increase in nNOS-positive fibers in the ileal muscle wall and a significant decrease in ChAT-positive myenteric neurons in GLP-2-exposed preparations. The present results offer the first evidence that GLP-2 acts on ileal preparations. The hormone appears to depress ileal contractility through a dual opposite modulatory effect on inhibitory nitrergic and excitatory cholinergic neurotransmission. From a physiological point of view, it could be hypothesized that GLP-2 inhibitory actions on ileal contractility can increase transit time, facilitating nutrient absorption.
Collapse
Affiliation(s)
- Eglantina Idrizaj
- Section of Physiological Sciences, Department of Experimental & Clinical Medicine, University of Florence, 50139 Florence, Italy;
| | - Cristina Biagioni
- Research Unit of Histology & Embryology, Department of Experimental & Clinical Medicine, University of Florence, 50139 Florence, Italy; (C.B.); (C.T.)
| | - Chiara Traini
- Research Unit of Histology & Embryology, Department of Experimental & Clinical Medicine, University of Florence, 50139 Florence, Italy; (C.B.); (C.T.)
| | - Maria Giuliana Vannucchi
- Research Unit of Histology & Embryology, Department of Experimental & Clinical Medicine, University of Florence, 50139 Florence, Italy; (C.B.); (C.T.)
| | - Maria Caterina Baccari
- Section of Physiological Sciences, Department of Experimental & Clinical Medicine, University of Florence, 50139 Florence, Italy;
| |
Collapse
|
11
|
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
|
12
|
Athavale ON, Avci R, Clark AR, Di Natale MR, Wang X, Furness JB, Liu Z, Cheng LK, Du P. Neural regulation of slow waves and phasic contractions in the distal stomach: a mathematical model. J Neural Eng 2024; 20:066040. [PMID: 38100816 PMCID: PMC10765034 DOI: 10.1088/1741-2552/ad1610] [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: 07/04/2023] [Revised: 11/06/2023] [Accepted: 12/15/2023] [Indexed: 12/17/2023]
Abstract
Objective.Neural regulation of gastric motility occurs partly through the regulation of gastric bioelectrical slow waves (SWs) and phasic contractions. The interaction of the tissues and organs involved in this regulatory process is complex. We sought to infer the relative importance of cellular mechanisms in inhibitory neural regulation of the stomach by enteric neurons and the interaction of inhibitory and excitatory electrical field stimulation.Approach.A novel mathematical model of gastric motility regulation by enteric neurons was developed and scenarios were simulated to determine the mechanisms through which enteric neural influence is exerted. This model was coupled to revised and extended electrophysiological models of gastric SWs and smooth muscle cells (SMCs).Main results.The mathematical model predicted that regulation of contractile apparatus sensitivity to intracellular calcium in the SMC was the major inhibition mechanism of active tension development, and that the effect on SW amplitude depended on the inhibition of non-specific cation currents more than the inhibition of calcium-activated chloride current (kiNSCC= 0.77 vs kiAno1= 0.33). The model predicted that the interaction between inhibitory and excitatory neural regulation, when applied with simultaneous and equal intensity, resulted in an inhibition of contraction amplitude almost equivalent to that of inhibitory stimulation (79% vs 77% decrease), while the effect on frequency was overall excitatory, though less than excitatory stimulation alone (66% vs 47% increase).Significance.The mathematical model predicts the effects of inhibitory and excitatory enteric neural stimulation on gastric motility function, as well as the effects when inhibitory and excitatory enteric neural stimulation interact. Incorporation of the model into organ-level simulations will provide insights regarding pathological mechanisms that underpin gastric functional disorders, and allow forin silicotesting of the effects of clinical neuromodulation protocols for the treatment of these disorders.
Collapse
Affiliation(s)
- Omkar N Athavale
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Recep Avci
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Madeleine R Di Natale
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Xiaokai Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - John B Furness
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Anatomy & Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Zhongming Liu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Leo K Cheng
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Peng Du
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
13
|
Xu Y, Liu RR, Yu XJ, Liu XN, Zhang X, Jiang ZH, Cong ZF, Li QQ, Gao P. Quality markers of Dajianzhong decoction based on multicomponent qualitative and quantitative analysis combined with network pharmacology and chemometric analysis. PHYTOCHEMICAL ANALYSIS : PCA 2024; 35:146-162. [PMID: 37731278 DOI: 10.1002/pca.3281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/20/2023] [Accepted: 08/20/2023] [Indexed: 09/22/2023]
Abstract
INTRODUCTION Dajianzhong decoction (DJZD), a classic famous prescription, has a long history of medicinal application. Modern studies have demonstrated its clinical utility in the treatment of postoperative ileus (POI). But none of the current quality evaluation methods for this compound is associated with efficacy. OBJECTIVES This study aimed to identify the quality markers (Q-Markers) connected to the treatment of POI in DJZD. METHODOLOGY Ultra-performance liquid chromatography quadrupole Exactive Orbitrap mass spectrometry (UPLC-Q-Exactive Orbitrap-MS) was used to identify the main constituents in DJZD. Based on the qualitative results obtained by fingerprinting, chemical pattern recognition (CPR) was used to analyse the key components affecting the quality and finally to establish the network of the active ingredients in DJZD with POI. RESULTS A total of 64 chemical components were detected. After fingerprint analysis, 13 common peaks were identified. The fingerprint similarity of 15 batches of samples ranged from 0.860 to 1.000. CPR analysis was able to categorically classify 15 batches of DJZD into two groups. And gingerenone A, methyl-6-gingerdiol, 6-gingerol, and hydroxy-β-sanshool contributed to their grouping. Twelve common components interact with the therapeutic targets for treating POI. In addition, the mechanism of this prescription for treating POI may be related to the jurisdiction of the neurological system, the immunological system, and the inflammatory response. CONCLUSIONS This integrated approach can accurately assess and forecast the quality of DJZD, presume the Q-Markers of DJZD for POI, and lay the foundation for studying the theoretical underpinnings and exploring the mechanism of DJZD in the treatment of POI.
Collapse
Affiliation(s)
- Yang Xu
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
- National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, P. R. China
| | - Run-Run Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Jun Yu
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Xiao-Nan Liu
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Xin Zhang
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Zhi-Hui Jiang
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
| | - Zhu-Feng Cong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, P. R. China
| | - Qin-Qing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Shanxi University of Chinese Medicine, Taiyuan, P. R. China
| | - Peng Gao
- Institute of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, P. R. China
- National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, P. R. China
| |
Collapse
|
14
|
Mazzoni M, Cabanillas L, Costanzini A, Caremoli F, Million M, Larauche M, Clavenzani P, De Giorgio R, Sternini C. Distribution, quantification, and characterization of substance P enteric neurons in the submucosal and myenteric plexuses of the porcine colon. Cell Tissue Res 2024; 395:39-51. [PMID: 37982872 PMCID: PMC10774220 DOI: 10.1007/s00441-023-03842-x] [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: 08/02/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023]
Abstract
The pig is an important translational model for studying intestinal physiology and disorders for its many homologies with humans, including the organization of the enteric nervous system (ENS), the major regulator of gastrointestinal functions. This study focused on the quantification and neurochemical characterization of substance P (SP) neurons in the pig ascending (AC) and descending colon (DC) in wholemount preparations of the inner submucosal plexus (ISP), outer submucosal plexus (OSP), and myenteric plexus (MP). We used antibodies for the pan-neuronal marker HuCD, and choline acetyltransferase (ChAT) and neuronal nitric oxide synthase (nNOS), markers for excitatory and inhibitory transmitters, for multiple labeling immunofluorescence and high-resolution confocal microscopy. The highest density of SP immunoreactive (IR) neurons was in the ISP (222/mm2 in the AC, 166/mm2 in the DC), where they make up about a third of HuCD-IR neurons, compared to the OSP and MP (19-22% and 13-17%, respectively, P < 0.001-0.0001). HuCD/SP/ChAT-IR neurons (up to 23%) were overall more abundant than HuCD/SP/nNOS-IR neurons (< 10%). Most SP-IR neurons contained ChAT-IR (62-85%), whereas 18-38% contained nNOS-IR with the highest peak in the OSP. A subpopulation of SP-IR neurons contains both ChAT- and nNOS-IR with the highest peak in the OSP and ISP of DC (33-36%) and the lowest in the ISP of AC (< 10%, P < 0.001). SP-IR varicose fibers were abundant in the ganglia. This study shows that SP-IR neurons are functionally distinct with variable proportions in different plexuses in the AC and DC reflecting diverse functions of specific colonic regions.
Collapse
Affiliation(s)
- Maurizio Mazzoni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, Bologna, Italy
| | - Luis Cabanillas
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Anna Costanzini
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Filippo Caremoli
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Current address: San Raffaele Hospital, Milan, Italy
| | - Mulugeta Million
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, 90095, USA
| | - Muriel Larauche
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Paolo Clavenzani
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, Bologna, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Catia Sternini
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
15
|
Wu Z, Wang Q, Yang F, Wang J, Zhao Y, Perrino BA, Chen J. Functional and Transcriptomic Characterization of Postnatal Maturation of ENS and SIP Syncytium in Mice Colon. Biomolecules 2023; 13:1688. [PMID: 38136560 PMCID: PMC10741935 DOI: 10.3390/biom13121688] [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: 08/22/2023] [Revised: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023] Open
Abstract
The interplay of the enteric nervous system (ENS) and SIP syncytium (smooth muscle cells-interstitial cells of Cajal-PDGFRα+ cells) plays an important role in the regulation of gastrointestinal (GI) motility. This study aimed to investigate the dynamic regulatory mechanisms of the ENS-SIP system on colon motility during postnatal development. Colonic samples of postnatal 1-week-old (PW1), 3-week-old (PW3), and 5-week-old (PW5) mice were characterized by RNA sequencing, qPCR, Western blotting, isometric force recordings (IFR), and colonic motor complex (CMC) force measurements. Our study showed that the transcriptional expression of Pdgfrα, c-Kit, P2ry1, Nos1, and Slc18a3, and the protein expression of nNOS, c-Kit, and ANO1 significantly increased with age from PW1 to PW5. In PW1 and PW3 mice, colonic migrating movement was not fully developed. In PW5 mice, rhythmic CMCs were recorded, similar to the CMC pattern described previously in adult mice. The inhibition of nNOS revealed excitatory and non-propulsive responses which are normally suppressed due to ongoing nitrergic inhibition. During postnatal development, molecular data demonstrated the establishment and expansion of ICC and PDGFRα+ cells, along with nitrergic and cholinergic nerves and purinergic receptors. Our findings are important for understanding the role of the SIP syncytium in generating and establishing CMCs in postnatal, developing murine colons.
Collapse
Affiliation(s)
- Zhihao Wu
- Department of General Surgery, Shanghai Children’s Medical Center, Shanghai Jiao Tong School of Medicine, Shanghai 200127, China
| | - Qianqian Wang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Fan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiaxuan Wang
- Department of General Surgery, Shanghai Children’s Medical Center, Shanghai Jiao Tong School of Medicine, Shanghai 200127, China
| | - Yuying Zhao
- Department of General Surgery, Shanghai Children’s Medical Center, Shanghai Jiao Tong School of Medicine, Shanghai 200127, China
| | - Brian A. Perrino
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Jie Chen
- Department of General Surgery, Shanghai Children’s Medical Center, Shanghai Jiao Tong School of Medicine, Shanghai 200127, China
| |
Collapse
|
16
|
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
|
17
|
Stavely R, Ott LC, Rashidi N, Sakkal S, Nurgali K. The Oxidative Stress and Nervous Distress Connection in Gastrointestinal Disorders. Biomolecules 2023; 13:1586. [PMID: 38002268 PMCID: PMC10669114 DOI: 10.3390/biom13111586] [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: 09/08/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress is increasingly recognized as a central player in a range of gastrointestinal (GI) disorders, as well as complications stemming from therapeutic interventions. This article presents an overview of the mechanisms of oxidative stress in GI conditions and highlights a link between oxidative insult and disruption to the enteric nervous system (ENS), which controls GI functions. The dysfunction of the ENS is characteristic of a spectrum of disorders, including neurointestinal diseases and conditions such as inflammatory bowel disease (IBD), diabetic gastroparesis, and chemotherapy-induced GI side effects. Neurons in the ENS, while essential for normal gut function, appear particularly vulnerable to oxidative damage. Mechanistically, oxidative stress in enteric neurons can result from intrinsic nitrosative injury, mitochondrial dysfunction, or inflammation-related pathways. Although antioxidant-based therapies have shown limited efficacy, recognizing the multifaceted role of oxidative stress in GI diseases offers a promising avenue for future interventions. This comprehensive review summarizes the literature to date implicating oxidative stress as a critical player in the pathophysiology of GI disorders, with a focus on its role in ENS injury and dysfunction, and highlights opportunities for the development of targeted therapeutics for these diseases.
Collapse
Affiliation(s)
- Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leah C. Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Niloufar Rashidi
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
- Department of Medicine Western Health, The University of Melbourne, St Albans, VIC 3021, Australia
- Regenerative Medicine and Stem Cell Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, VIC 3021, Australia
| |
Collapse
|
18
|
Choi EL, Taheri N, Tan E, Matsumoto K, Hayashi Y. The Crucial Role of the Interstitial Cells of Cajal in Neurointestinal Diseases. Biomolecules 2023; 13:1358. [PMID: 37759758 PMCID: PMC10526372 DOI: 10.3390/biom13091358] [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: 08/11/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Neurointestinal diseases result from dysregulated interactions between the nervous system and the gastrointestinal (GI) tract, leading to conditions such as Hirschsprung's disease and irritable bowel syndrome. These disorders affect many people, significantly diminishing their quality of life and overall health. Central to GI motility are the interstitial cells of Cajal (ICC), which play a key role in muscle contractions and neuromuscular transmission. This review highlights the role of ICC in neurointestinal diseases, revealing their association with various GI ailments. Understanding the functions of the ICC could lead to innovative perspectives on the modulation of GI motility and introduce new therapeutic paradigms. These insights have the potential to enhance efforts to combat neurointestinal diseases and may lead to interventions that could alleviate or even reverse these conditions.
Collapse
Affiliation(s)
- Egan L. Choi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Negar Taheri
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Elijah Tan
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Kenjiro Matsumoto
- Laboratory of Pathophysiology, Faculty of Pharmaceutical Sciences, Doshisha Women’s College of Liberal Arts, Kyoto 610-0395, Japan;
| | - Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
19
|
Mawe GM, Sanders KM, Camilleri M. Overview of the Enteric Nervous System. Semin Neurol 2023; 43:495-505. [PMID: 37562453 DOI: 10.1055/s-0043-1771466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Propulsion of contents in the gastrointestinal tract requires coordinated functions of the extrinsic nerves to the gut from the brain and spinal cord, as well as the neuromuscular apparatus within the gut. The latter includes excitatory and inhibitory neurons, pacemaker cells such as the interstitial cells of Cajal and fibroblast-like cells, and smooth muscle cells. Coordination between these extrinsic and enteric neurons results in propulsive functions which include peristaltic reflexes, migrating motor complexes in the small intestine which serve as the housekeeper propelling to the colon the residual content after digestion, and mass movements in the colon which lead to defecation.
Collapse
Affiliation(s)
- Gary M Mawe
- Department of Neurological Sciences, The University of Vermont, Burlington, Vermont
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
20
|
Buonfiglio F, Böhm EW, Pfeiffer N, Gericke A. Oxidative Stress: A Suitable Therapeutic Target for Optic Nerve Diseases? Antioxidants (Basel) 2023; 12:1465. [PMID: 37508003 PMCID: PMC10376185 DOI: 10.3390/antiox12071465] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Optic nerve disorders encompass a wide spectrum of conditions characterized by the loss of retinal ganglion cells (RGCs) and subsequent degeneration of the optic nerve. The etiology of these disorders can vary significantly, but emerging research highlights the crucial role of oxidative stress, an imbalance in the redox status characterized by an excess of reactive oxygen species (ROS), in driving cell death through apoptosis, autophagy, and inflammation. This review provides an overview of ROS-related processes underlying four extensively studied optic nerve diseases: glaucoma, Leber's hereditary optic neuropathy (LHON), anterior ischemic optic neuropathy (AION), and optic neuritis (ON). Furthermore, we present preclinical findings on antioxidants, with the objective of evaluating the potential therapeutic benefits of targeting oxidative stress in the treatment of optic neuropathies.
Collapse
Affiliation(s)
- Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (E.W.B.); (N.P.)
| | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (E.W.B.); (N.P.)
| |
Collapse
|
21
|
Chen BN, Humenick A, Yew WP, Peterson RA, Wiklendt L, Dinning PG, Spencer NJ, Wattchow DA, Costa M, Brookes SJH. Types of Neurons in the Human Colonic Myenteric Plexus Identified by Multilayer Immunohistochemical Coding. Cell Mol Gastroenterol Hepatol 2023; 16:573-605. [PMID: 37355216 PMCID: PMC10469081 DOI: 10.1016/j.jcmgh.2023.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND AIMS Gut functions including motility, secretion, and blood flow are largely controlled by the enteric nervous system. Characterizing the different classes of enteric neurons in the human gut is an important step to understand how its circuitry is organized and how it is affected by disease. METHODS Using multiplexed immunohistochemistry, 12 discriminating antisera were applied to distinguish different classes of myenteric neurons in the human colon (2596 neurons, 12 patients) according to their chemical coding. All antisera were applied to every neuron, in multiple layers, separated by elutions. RESULTS A total of 164 combinations of immunohistochemical markers were present among the 2596 neurons, which could be divided into 20 classes, with statistical validation. Putative functions were ascribed for 4 classes of putative excitatory motor neurons (EMN1-4), 4 inhibitory motor neurons (IMN1-4), 3 ascending interneurons (AIN1-3), 6 descending interneurons (DIN1-6), 2 classes of multiaxonal sensory neurons (SN1-2), and a small, miscellaneous group (1.8% of total). Soma-dendritic morphology was analyzed, revealing 5 common shapes distributed differentially between the 20 classes. Distinctive baskets of axonal varicosities surrounded 45% of myenteric nerve cell bodies and were associated with close appositions, suggesting possible connectivity. Baskets of cholinergic terminals and several other types of baskets selectively targeted ascending interneurons and excitatory motor neurons but were significantly sparser around inhibitory motor neurons. CONCLUSIONS Using a simple immunohistochemical method, human myenteric neurons were shown to comprise multiple classes based on chemical coding and morphology and dense clusters of axonal varicosities were selectively associated with some classes.
Collapse
Affiliation(s)
- Bao Nan Chen
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Adam Humenick
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Rochelle A Peterson
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Lukasz Wiklendt
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia; Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Nick J Spencer
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - David A Wattchow
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Marcello Costa
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Simon J H Brookes
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
22
|
López-Pingarrón L, Almeida H, Soria-Aznar M, Reyes-Gonzales MC, Rodríguez-Moratinos AB, Muñoz-Hoyos A, García JJ. Interstitial Cells of Cajal and Enteric Nervous System in Gastrointestinal and Neurological Pathology, Relation to Oxidative Stress. Curr Issues Mol Biol 2023; 45:3552-3572. [PMID: 37185756 PMCID: PMC10136929 DOI: 10.3390/cimb45040232] [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: 03/11/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
The enteric nervous system (ENS) is organized into two plexuses-submucosal and myenteric-which regulate smooth muscle contraction, secretion, and blood flow along the gastrointestinal tract under the influence of the rest of the autonomic nervous system (ANS). Interstitial cells of Cajal (ICCs) are mainly located in the submucosa between the two muscle layers and at the intramuscular level. They communicate with neurons of the enteric nerve plexuses and smooth muscle fibers and generate slow waves that contribute to the control of gastrointestinal motility. They are also involved in enteric neurotransmission and exhibit mechanoreceptor activity. A close relationship appears to exist between oxidative stress and gastrointestinal diseases, in which ICCs can play a prominent role. Thus, gastrointestinal motility disorders in patients with neurological diseases may have a common ENS and central nervous system (CNS) nexus. In fact, the deleterious effects of free radicals could affect the fine interactions between ICCs and the ENS, as well as between the ENS and the CNS. In this review, we discuss possible disturbances in enteric neurotransmission and ICC function that may cause anomalous motility in the gut.
Collapse
Affiliation(s)
- Laura López-Pingarrón
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Henrique Almeida
- i3S-Instituto de Investigação e Inovação em Saúde, Porto University, 4200-135 Porto, Portugal
- Department of Biomedicine, Faculty of Medicine, Porto University, 4200-319 Porto, Portugal
- Department of Obstetrics and Gynecology, Hospital-CUF Porto, 4100-180 Porto, Portugal
| | - Marisol Soria-Aznar
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Marcos C Reyes-Gonzales
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | | | - Antonio Muñoz-Hoyos
- Department of Pediatrics, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Joaquín J García
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| |
Collapse
|
23
|
Vijay A, Boyle NR, Kumar SM, Perdew GH, Srinivasan S, Patterson AD. Aryl hydrocarbon receptor activation affects nitrergic neuronal survival and delays intestinal motility in mice. Toxicol Sci 2023; 192:117-128. [PMID: 36782369 PMCID: PMC10025877 DOI: 10.1093/toxsci/kfad014] [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] [Indexed: 02/15/2023] Open
Abstract
Despite progress describing the effects of persistent organic pollutants (POPs) on the central nervous system, the effect of POPs on enteric nervous system (ENS) function remains underexplored. We studied the effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a POP, and a potent aryl hydrocarbon receptor (AHR) ligand, on the ENS and intestinal motility in mice. C57Bl/6J mice treated with TCDD (2.4 µg/kg body weight) for 8 weeks (once per week) exhibited significant delay in intestinal motility as shown by reduced stool frequency, prolonged intestinal transit time, and a persistence of dye in the jejunum compared to control mice with maximal dye retention in the ileum. TCDD significantly increased Cyp1a1 expression, an AHR target gene, and reduced the total number of neurons and affected nitrergic neurons in cells isolated from WT mice, but not Ahr-/- mice. In immortalized fetal enteric neuronal cells, TCDD-induced nuclear translocation of AHR as well as increased Cyp1a1 expression. AHR activation did not affect neuronal proliferation. However, AHR activation resulted in enteric neuronal toxicity, specifically, nitrergic neurons. Our results demonstrate that TCDD adversely affects nitrergic neurons and thereby contributes to delayed intestinal motility. These findings suggest that AHR signaling in the ENS may play a role in modulating TCDD-induced gastrointestinal pathophysiology.
Collapse
Affiliation(s)
- Anitha Vijay
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Nina R Boyle
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Supriya M Kumar
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Gary H Perdew
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Shanthi Srinivasan
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
24
|
Athavale ON, Avci R, Cheng LK, Du P. Computational models of autonomic regulation in gastric motility: Progress, challenges, and future directions. Front Neurosci 2023; 17:1146097. [PMID: 37008202 PMCID: PMC10050371 DOI: 10.3389/fnins.2023.1146097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
The stomach is extensively innervated by the vagus nerve and the enteric nervous system. The mechanisms through which this innervation affects gastric motility are being unraveled, motivating the first concerted steps towards the incorporation autonomic regulation into computational models of gastric motility. Computational modeling has been valuable in advancing clinical treatment of other organs, such as the heart. However, to date, computational models of gastric motility have made simplifying assumptions about the link between gastric electrophysiology and motility. Advances in experimental neuroscience mean that these assumptions can be reviewed, and detailed models of autonomic regulation can be incorporated into computational models. This review covers these advances, as well as a vision for the utility of computational models of gastric motility. Diseases of the nervous system, such as Parkinson’s disease, can originate from the brain-gut axis and result in pathological gastric motility. Computational models are a valuable tool for understanding the mechanisms of disease and how treatment may affect gastric motility. This review also covers recent advances in experimental neuroscience that are fundamental to the development of physiology-driven computational models. A vision for the future of computational modeling of gastric motility is proposed and modeling approaches employed for existing mathematical models of autonomic regulation of other gastrointestinal organs and other organ systems are discussed.
Collapse
|
25
|
Oh HK, Sung TS, Ryoo SB, Park KJ. Regional Differences in Intestinal Contractile Responses to Radial Stretch in the Human Lower Gastrointestinal Tract. J Neurogastroenterol Motil 2023; 29:113-121. [PMID: 36437512 PMCID: PMC9837542 DOI: 10.5056/jnm21236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/01/2022] [Accepted: 08/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background/Aims Radial stretch evokes an increase or decrease in contractions in the lower gastrointestinal tract via mechanosensory enteric neurons that project into the muscle layers. We aim to elucidate the differences in stretch reflexes according to their location in the human colon. Methods We used healthy intestinal smooth muscle tissue excised during elective colon cancer surgery. Conventional intracellular recordings from colonic muscle cells and tension recordings of colonic segments were performed. Radial stretch was evoked through balloon catheter inflation. Changes in the membrane potential and frequency, amplitude, and area under the curve of muscle contractions were recorded before and after the radial stretch at proximal and distal segment sites. Results In intracellular circular muscle recordings, hyperpolarization was noted at the distal site of sigmoid colonic segments after radial stretch, in contrast to depolarization at all other sites. In tension recordings at proximal ascending or sigmoid colonic segment sites, contractile activation was observed with statistically significant increases in the frequency, amplitude, and area under the curve after radial stretch. Distal sites of ascending and sigmoid colonic segments showed increase and decrease in contraction, respectively. Conclusion Radial stretch in the human colon (in vitro) evokes excitatory activity at both proximal and distal sites of the ascending colon and at the proximal site of the sigmoid colon, whereas it elicits inhibitory activity at the distal site of the sigmoid colon.
Collapse
Affiliation(s)
- Heung-Kwon Oh
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Tae Sik Sung
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyu Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea,Correspondence: Kyu Joo Park, MD, PhD, Department of Surgery, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, Seoul 03080, Korea, Tel: +82-2-2072-2901, Fax: +82-2-766-3975, E-mail:
| |
Collapse
|
26
|
Xue M, Cao Y, Shen C, Guo W. Computational Advances of Protein/Neurotransmitter-membrane Interactions Involved in Vesicle Fusion and Neurotransmitter Release. J Mol Biol 2023; 435:167818. [PMID: 36089056 DOI: 10.1016/j.jmb.2022.167818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/22/2022] [Accepted: 09/04/2022] [Indexed: 02/04/2023]
Abstract
Vesicle fusion is of crucial importance to neuronal communication at neuron terminals. The exquisite but complex fusion machinery for neurotransmitter release is tightly controlled and regulated by protein/neurotransmitter-membrane interactions. Computational 'microscopies', in particular molecular dynamics simulations and related techniques, have provided notable insight into the physiological process over the past decades, and have made enormous contributions to fields such as neurology, pharmacology and pathophysiology. Here we review the computational advances of protein/neurotransmitter-membrane interactions related to presynaptic vesicle-membrane fusion and neurotransmitter release, and outline the in silico challenges ahead for understanding this important physiological process.
Collapse
Affiliation(s)
- Minmin Xue
- Key Laboratory for Intelligent Nano Materials and Devices of Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, and Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Yuwei Cao
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Chun Shen
- Key Laboratory for Intelligent Nano Materials and Devices of Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, and Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China.
| | - Wanlin Guo
- Key Laboratory for Intelligent Nano Materials and Devices of Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, and Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, China.
| |
Collapse
|
27
|
Guo B, Huang X, Chen Y, Broxmeyer HE. Ex Vivo Expansion and Homing of Human Cord Blood Hematopoietic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:85-104. [PMID: 38228960 DOI: 10.1007/978-981-99-7471-9_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Cord blood (CB) has been proven to be an alternative source of haematopoietic stem cells (HSCs) for clinical transplantation and has multiple advantages, including but not limited to greater HLA compatibility, lower incidence of graft-versus-host disease (GvHD), higher survival rates and lower relapse rates among patients with minimal residual disease. However, the limited number of HSCs in a single CB unit limits the wider use of CB in clinical treatment. Many efforts have been made to enhance the efficacy of CB HSC transplantation, particularly by ex vivo expansion or enhancing the homing efficiency of HSCs. In this chapter, we will document the major advances regarding human HSC ex vivo expansion and homing and will also discuss the possibility of clinical translation of such laboratory work.
Collapse
Affiliation(s)
- Bin Guo
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xinxin Huang
- Xuhui Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Yandan Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, School of Medicine, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
28
|
The Comparison of the Influence of Bisphenol A (BPA) and Its Analogue Bisphenol S (BPS) on the Enteric Nervous System of the Distal Colon in Mice. Nutrients 2022; 15:nu15010200. [PMID: 36615857 PMCID: PMC9824883 DOI: 10.3390/nu15010200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Bisphenol A (BPA), commonly used as a plasticizer in various branches of industry has a strong negative effect on living organisms. Therefore, more and more often it is replaced in production of plastics by other substances. One of them is bisphenol S (BPS). This study for the first time compares the impact of BPA and BPS on the enteric neurons using double immunofluorescence technique. It has been shown that both BPA and BPS affect the number of enteric neurons containing substance P (SP), galanin (GAL), vasoactive intestinal polypeptide (VIP), neuronal isoform of nitric oxide synthase (nNOS-a marker of nitrergic neurons) and/or vesicular acetylcholine transporter (VAChT- a marker of cholinergic neurons). The changes noted under the impact of both bisphenols are similar and consisted of an increase in the number of enteric neurons immunoreactive to all neuronal factors studied. The impact of BPS on some populations of neurons was stronger than that noted under the influence of BPA. The obtained results clearly show that BPS (similarly to BPA) administered for long time is not neutral for the enteric neurons even in relatively low doses and may be more potent than BPA for certain neuronal populations.
Collapse
|
29
|
Neuronal Nitric Oxide Synthase as a Shared Target for the Effects of Adiponectin and Resistin on the Mechanical Responses of the Mouse Gastric Fundus. Int J Mol Sci 2022; 23:ijms232416113. [PMID: 36555750 PMCID: PMC9781802 DOI: 10.3390/ijms232416113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
It has been reported that adiponectin (ADPN) and resistin are co-secreted by white mouse adipocytes and exert similar inhibitory effects in the mouse gastric fundus, in which resistin was observed to increase neuronal nitric oxide synthase (nNOS) expression. On these grounds, the present work aimed to investigate whether the effects of the two adipokines on the neurally-induced relaxant responses potentiate each other and whether there is a possible correlation with changes in nNOS expression in preparations from the mouse gastric fundus. In carbachol (CCh)-precontracted strips, electrical field stimulation elicited nitrergic relaxant responses, whose amplitude was increased by ADPN or resistin, but no additional enhancements were observed in their concomitant presence. Western blot and immunofluorescence analyses revealed that ADPN, like resistin, was able to up-regulate nNOS expression and to increase the percentage of nNOS-positive neurons in the myenteric plexus: co-treatment with the two adipokines did not induce additional changes. The results indicate that the two adipokines modulate nitrergic neurotransmission, and both do so by up-regulating nNOS expression. Therefore, nNOS appears to be a shared target for the two adipokines' effects, which, rather than mutually reinforcing each other, may represent a dual physiological control mechanism to guarantee gastric fundus relaxation.
Collapse
|
30
|
Peterson JAM, Cooper TA. Clinical and Molecular Insights into Gastrointestinal Dysfunction in Myotonic Dystrophy Types 1 & 2. Int J Mol Sci 2022; 23:ijms232314779. [PMID: 36499107 PMCID: PMC9737721 DOI: 10.3390/ijms232314779] [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: 10/25/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Myotonic dystrophy (DM) is a highly variable, multisystemic disorder that clinically affects one in 8000 individuals. While research has predominantly focused on the symptoms and pathological mechanisms affecting striated muscle and brain, DM patient surveys have identified a high prevalence for gastrointestinal (GI) symptoms amongst affected individuals. Clinical studies have identified chronic and progressive dysfunction of the esophagus, stomach, liver and gallbladder, small and large intestine, and rectum and anal sphincters. Despite the high incidence of GI dysmotility in DM, little is known regarding the pathological mechanisms leading to GI dysfunction. In this review, we summarize results from clinical and molecular analyses of GI dysfunction in both genetic forms of DM, DM type 1 (DM1) and DM type 2 (DM2). Based on current knowledge of DM primary pathological mechanisms in other affected tissues and GI tissue studies, we suggest that misregulation of alternative splicing in smooth muscle resulting from the dysregulation of RNA binding proteins muscleblind-like and CUGBP-elav-like is likely to contribute to GI dysfunction in DM. We propose that a combinatorial approach using clinical and molecular analysis of DM GI tissues and model organisms that recapitulate DM GI manifestations will provide important insight into defects impacting DM GI motility.
Collapse
Affiliation(s)
- Janel A. M. Peterson
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor College of Medicine, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas A. Cooper
- Baylor College of Medicine, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor College of Medicine, Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor College of Medicine, Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
31
|
Involvement of NO/cGMP Signaling Pathway, Ca 2+ and K + Channels on Spasmolytic Effect of Everlasting Flower Polyphenolic Extract ( Helichrysum stoechas (L.) Moench). Int J Mol Sci 2022; 23:ijms232214422. [PMID: 36430899 PMCID: PMC9696489 DOI: 10.3390/ijms232214422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Functional gastrointestinal diseases (FGID) are worldwide prevalent conditions. Pharmacological treatments can be ineffective, leading the population to turn to herbal or traditional remedies. Helichrysum stoechas (L.) Moench is a medicinal plant traditionally used in the Iberian Peninsula to treat digestive disorders, but its effects on gastrointestinal motility have not been scientifically demonstrated. The aim of this work was to evaluate the antispasmodic effect of a polyphenolic extract of H. stoechas (HSM), its mechanism of action and its antioxidant activity. Isometric myography studies were performed in rat ileum, and malondialdehyde (MDA) and 4-hydroxyalkenals (4-HDA) levels were measured in rat jejunum. HSM reduced the integrated mechanical activity of spontaneous contractions. In Ca2+-free medium, HSM reduced the concentration-response curve of CaCl2 similarly to verapamil. Pre-incubation with the extract blocked the contraction induced by Bay K8644, KCl and carbachol. L-NAME, ODQ, Rp-8-Br-PET-cGMPS, KT-5823, apamin, TRAM-34 and charybdotoxin reduced the relaxant effect of the extract on spontaneous contractions. MDA+4-HDA levels in LPS-treated tissue were reduced by the extract, showing antioxidant activity. In conclusion, HSM showed antispasmodic activity through inhibition of Ca2+ influx, activation of the NO/PKG/cGMP pathway and opening of Ca2+-activated K+ channels. The results suggest that H. stoechas could help in the prevention or treatment of FGIDs.
Collapse
|
32
|
Grabacka M, Płonka PM, Pierzchalska M. The PPARα Regulation of the Gut Physiology in Regard to Interaction with Microbiota, Intestinal Immunity, Metabolism, and Permeability. Int J Mol Sci 2022; 23:ijms232214156. [PMID: 36430628 PMCID: PMC9696208 DOI: 10.3390/ijms232214156] [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: 10/27/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) is expressed throughout the mammalian gut: in epithelial cells, in the villi of enterocytes and in Paneth cells of intestinal crypts, as well as in some immune cells (e.g., lamina propria macrophages, dendritic cells) of the mucosa. This review examines the reciprocal interaction between PPARα activation and intestinal microbiota. We refer to the published data confirming that microbiota products can influence PPARα signaling and, on the other hand, PPARα activation is able to affect microbiota profile, viability, and diversity. PPARα impact on the broad spectrum of events connected to metabolism, signaling (e.g., NO production), immunological tolerance to dietary antigens, immunity and permeability of the gut are also discussed. We believe that the phenomena described here play a prominent role in gut homeostasis. Therefore, in conclusion we propose future directions for research, including the application of synthetic activators and natural endogenous ligands of PPARα (i.e., endocannabinoids) as therapeutics for intestinal pathologies and systemic diseases assumed to be related to gut dysbiosis.
Collapse
Affiliation(s)
- Maja Grabacka
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, ul. Balicka 122, 30-149 Cracow, Poland
- Correspondence: ; Tel.: +48-12-662-4701
| | - Przemysław M. Płonka
- Department of Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Cracow, Poland
| | - Małgorzata Pierzchalska
- Department of Biotechnology and General Technology of Foods, Faculty of Food Technology, University of Agriculture, ul. Balicka 122, 30-149 Cracow, Poland
| |
Collapse
|
33
|
Fan M, Shi H, Yao H, Wang W, Zhang Y, Jiang C, Lin R. BMSCs Promote Differentiation of Enteric Neural Precursor Cells to Maintain Neuronal Homeostasis in Mice With Enteric Nerve Injury. Cell Mol Gastroenterol Hepatol 2022; 15:511-531. [PMID: 36343901 PMCID: PMC9880979 DOI: 10.1016/j.jcmgh.2022.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND & AIMS Our previous study showed that transplantation of bone marrow-derived mesenchymal stem cells (BMSCs) promoted functional enteric nerve regeneration in denervated mice but not through direct transdifferentiation. Homeostasis of the adult enteric nervous system (ENS) is maintained by enteric neural precursor cells (ENPCs). Whether ENPCs are a source of regenerated nerves in denervated mice remains unknown. METHODS Genetically engineered mice were used as recipients, and ENPCs were traced during enteric nerve regeneration. The mice were treated with benzalkonium chloride to establish a denervation model and then transplanted with BMSCs 3 days later. After 28 days, the gastric motility and ENS regeneration were analyzed. The interaction between BMSCs and ENPCs in vitro was further assessed. RESULTS Twenty-eight days after transplantation, gastric motility recovery (gastric emptying capacity, P < .01; gastric contractility, P < .01) and ENS regeneration (neurons, P < .01; glial cells, P < .001) were promoted in BMSCs transplantation groups compared with non-transplanted groups in denervated mice. More importantly, we found that ENPCs could differentiate into enteric neurons and glial cells in denervated mice after BMSCs transplantation, and the proportion of Nestin+/Ngfr+ cells differentiated into neurons was significantly higher than that of Nestin+ cells. A small number of BMSCs located in the myenteric plexus differentiated into glial cells. In vitro, glial cell-derived neurotrophic factor (GDNF) from BMSCs promotes the migration, proliferation, and differentiation of ENPCs. CONCLUSIONS In the case of enteric nerve injury, ENPCs can differentiate into enteric neurons and glial cells to promote ENS repair and gastric motility recovery after BMSCs transplantation. BMSCs expressing GDNF enhance the migration, proliferation, and differentiation of ENPCs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rong Lin
- Correspondence Address correspondence to: Rong Lin, MD, PhD, Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
34
|
Hwang SJ, Drumm BT, Kim MK, Lyu JH, Baker S, Sanders KM, Ward SM. Calcium transients in intramuscular interstitial cells of Cajal of the murine gastric fundus and their regulation by neuroeffector transmission. J Physiol 2022; 600:4439-4463. [PMID: 36057845 DOI: 10.1113/jp282876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/15/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The cells responsible for mediating enteric neuroeffector transmission remain controversial. In the stomach intramuscular interstitial cells of Cajal (ICC-IM) were the first ICC reported to receive cholinergic and nitrergic neural inputs. Utilization of a cell specific calcium biosensor, GCaMP6f, the activity and neuroeffector responses of ICC-IM were examined. ICC-IM were highly active, generating stochastic intracellular Ca2+ -transients. Stimulation of enteric motor nerves abolished Ca2+ -transients in ICC-IM. This inhibitory response was preceded by a global rise in intracellular Ca2+ . Individual ICC-IM responded to nerve stimulation with a rise in Ca2+ followed by inhibition of Ca2+ -transients. Inhibition of Ca2+ -transients was blocked by the nitric oxide synthase antagonist, L-NNA. The global rise in Ca2+ was inhibited by the muscarinic antagonist, atropine. Simultaneous intracellular recordings with video imaging revealed that the global rise in intracellular Ca2+ and inhibition of Ca2+ -transients was temporally associated with rapid excitatory junction potentials followed by more sustained inhibitory junction potentials. The data presented support the premise of serial innervation of ICC-IM in excitatory and inhibitory neuroeffector transmission in the proximal stomach. ABSTRACT Enteric neurotransmission is critical for coordinating motility throughout the gastrointestinal (GI) tract. However, there is considerable controversy regarding the cells that are responsible for the transduction of these neural inputs. In the present study, utilization of a cell-specific calcium biosensor GCaMP6f, the spontaneous activity and neuroeffector responses of intramuscular ICC (ICC-IM) to motor neural inputs was examined. Simultaneous intracellular microelectrode recordings and high-speed video-imaging during nerve stimulation was used to reveal the temporal relationship between changes in intracellular Ca2+ and post-junctional electrical responses to neural stimulation. ICC-IM were highly active, generating intracellular Ca2+ -transients that occurred stochastically, from multiple independent sites in single ICC-IM. Ca2+ -transients were not entrained in single ICC-IM or between neighboring ICC-IM. Activation of enteric motor neurons produced a dominant inhibitory response that abolished Ca2+ -transients in ICC-IM. This inhibitory response was often preceded by a summation of Ca2+ -transients that led to a global rise in Ca2+ . Individual ICC-IM responded to nerve stimulation by a global rise in Ca2+ followed by inhibition of Ca2+ -transients. The inhibition of Ca2+ -transients was blocked by the nitric oxide synthase antagonist, L-NNA. The global rise in intracellular Ca2+ was inhibited by the muscarinic antagonist, atropine. Simultaneous intracellular microelectrode recordings with video-imaging revealed that the rise in Ca2+ was temporally associated with rapid excitatory junction potentials and the inhibition of Ca2+ -transients with inhibitory junction potentials. These data support the premise of serial innervation of ICC-IM in excitatory and inhibitory neuroeffector transmission in the proximal stomach. Abstract figure legend Intramuscular interstitial cells of Cajal (ICC-IM) of the gastric fundus receive nitrergic inhibitory and cholinergic excitatory neuroeffector motor inputs. Using a genetically encoded calcium sensor we demonstrate that ICC-IM are highly active cells generating stochastic intracellular Ca2 -transients. Stimulation of enteric motor nerves abolished Ca2 -transients in ICC-IM, produced an inhibitory junction potential (IJP) and muscle relaxation that was mediated by nitric oxide (left hand side of figure). This inhibitory response was often preceded by a global rise in intracellular Ca2 in ICC-IM, a rapid excitatory junction potential (EJP) and muscle contraction, that was mediated by acetylcholine (right hand side of figure). Individual ICC-IM could respond to both excitatory and inhibitory neural inputs. These data support the premise of serial innervation of ICC-IM in excitatory and inhibitory neuroeffector transmission in the proximal stomach. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Bernard T Drumm
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Min Kyung Kim
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Ju Hyeong Lyu
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sal Baker
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| |
Collapse
|
35
|
Lundberg JO, Weitzberg E. Nitric oxide signaling in health and disease. Cell 2022; 185:2853-2878. [DOI: 10.1016/j.cell.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 10/16/2022]
|
36
|
Filho FC, Silva JDP, Petri C, Almendra JSL, de Sousa ÍA, Cavalcanti SMG, Silva BA, Formiga Melo MF, Cavalcanti PMDS. Pharmacological evidence that GABA-induced relaxation of rat proximal duodenum longitudinal muscle depends on NKCC cotransporter activity and Ca 2+ influx. Can J Physiol Pharmacol 2022; 100:728-740. [PMID: 35880679 DOI: 10.1139/cjpp-2021-0639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in adult central nervous system (CNS) synapses, but it excites immature CNS neurons as well as neurons in the myenteric plexus. The present work aimed to determine whether GABA-induced nonadrenergic, noncholinergic (NANC) neuronal-mediated relaxation of the rat duodenum is dependent on the activity of Na+ K+ Cl- cotransporters (NKCC) and requires calcium influx. In the presence of guanethidine (3 µmol/L), atropine (3 µmol/L), and indomethacin (1 µmol/L), relaxations induced by GABA (100 µmol/L), KCl (5-10 mmol/L) and electrical field stimulation (1-8 Hz, 2 ms, 60 V), but not those induced by bradykinin (10-100 nmol/L) were abolished by lidocaine (300 µmol/L). However, only GABA-induced relaxations were reduced in a concentration-dependent manner by the NKCC1/2 inhibitors bumetanide (0.1-1 µmol/L) and furosemide (1-10 µmol/L). GABA-induced NANC neuronal relaxation was abolished by bicuculline (30 µmol/L) and inhibited by N-nitroarginine methyl ester (l-NAME, 300 µmol/L). The ω-conotoxin GVIA (1 µmol/L), which acts exclusively on neuronal CaV2 channels, but not on smooth muscle voltage-gated Ca2+ CaV1 channels, and nonselective blockers of these channels (verapamil 100 nmol/L and ruthenium red 10 µmol/L), reduced GABA-induced relaxations. These results showed that the activation of GABAA receptors induces NANC nitrergic neuronal relaxations in the rat duodenum, which depend on NKCC activity and CaV2 channel activation, suggesting that this phenomenon results from neuronal depolarization promoted by Cl- efflux through GABAA receptors, with subsequent Ca2+ influx and nitric oxide release.
Collapse
Affiliation(s)
- Francisco Chagas Filho
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Janyerson Dannys Pereira Silva
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Caio Petri
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - João Santos Lima Almendra
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Ícaro Araújo de Sousa
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | - Bagnólia A Silva
- Pharmacological Sciences Department, 58051-900, Health Sciences Center, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Margareth Fátima Formiga Melo
- Pharmacological Sciences Department, 58051-900, Health Sciences Center, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | |
Collapse
|
37
|
Idrizaj E, Garella R, Nistri S, Squecco R, Baccari MC. Evidence that resistin acts on the mechanical responses of the mouse gastric fundus. Front Physiol 2022; 13:930197. [PMID: 35910552 PMCID: PMC9334560 DOI: 10.3389/fphys.2022.930197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Resistin, among its several actions, has been reported to exert central anorexigenic effects in rodents. Some adipokines which centrally modulate food intake have also been reported to affect the activity of gastric smooth muscle, whose motor responses represent a source of peripheral signals implicated in the control of the hunger-satiety cycle through the gut-brain axis. On this basis, in the present experiments, we investigated whether resistin too could affect the mechanical responses in the mouse longitudinal gastric fundal strips. Electrical field stimulation (EFS) elicited tetrodotoxin- and atropine-sensitive contractile responses. Resistin reduced the amplitude of the EFS-induced contractile responses. This effect was no longer detected in the presence of L-NNA, a nitric oxide (NO) synthesis inhibitor. Resistin did not influence the direct muscular response to methacholine. In the presence of carbachol and guanethidine, EFS elicited inhibitory responses whose amplitude was increased by resistin. L-NNA abolished the inhibitory responses evoked by EFS, indicating their nitrergic nature. In the presence of L-NNA, resistin did not have any effect on the EFS-evoked inhibitory responses. Western blot and immunofluorescence analysis revealed a significant increase in neuronal nitric oxide synthase (nNOS) expression in neurons of the myenteric plexus following resistin exposure. In conclusion, the present results offer the first evidence that resistin acts on the gastric fundus, likely through a modulatory action on the nitrergic neurotransmission.
Collapse
Affiliation(s)
- Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
- *Correspondence: Eglantina Idrizaj, ; Maria Caterina Baccari,
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Silvia Nistri
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Maria Caterina Baccari
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
- *Correspondence: Eglantina Idrizaj, ; Maria Caterina Baccari,
| |
Collapse
|
38
|
Prohibitins: A Key Link between Mitochondria and Nervous System Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7494863. [PMID: 35847581 PMCID: PMC9286927 DOI: 10.1155/2022/7494863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 06/21/2022] [Indexed: 12/02/2022]
Abstract
Prohibitins (PHBs) are conserved proteins in eukaryotic cells, which are mainly located in the inner mitochondrial membrane (IMM), cell nucleus, and cell membrane. PHBs play crucial roles in various cellular functions, including the cell cycle regulation, tumor suppression, immunoglobulin M receptor binding, and aging. In addition, recent in vitro and in vivo studies have revealed that PHBs are important in nervous system diseases. PHBs can prevent apoptosis, inflammation, mitochondrial dysfunction, and autophagy in neurological disorders through different molecules and pathways, such as OPA-1, PINK1/Parkin, IL6/STAT3, Tau, NO, LC3, and TDP43. Therefore, PHBs show great promise in the protection of neurological disorders. This review summarizes the relevant studies on the relationship between PHBs and neurological disorders and provides an update on the molecular mechanisms of PHBs in nervous system diseases.
Collapse
|
39
|
Athavale ON, Cheng LK, Clark AR, Avci R, Du P. Mathematical Modeling of Gastric Slow Waves During Electrical Field Stimulation. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:2266-2269. [PMID: 36086185 DOI: 10.1109/embc48229.2022.9871307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
While neural modulation has been trialed as a therapy for functional gastric motility disorders, a computational model that guides stimulation protocol does not exist. In this work, a mathematical model of gastric slow wave activity, which incorporates the effects of neurotransmitter release during electrical field stimulation (EFS), was developed. Slow wave frequency responses due to the release of acetylcholine and slow wave amplitude responses due to the release of nitric oxide were modeled. The model was calibrated using experimental data from literature. A sensitivity analysis was conducted, which showed that the model yielded stable, periodic solutions for EFS frequencies in the range 0-20 Hz. A 25% increase in the input parameter (EFS frequency) from 5 Hz to 6.25 Hz resulted in a 5.2% increase in slow wave frequency and a 3.2 % decrease in slow wave amplitude. Simulated EFS showed that, for stimulation at 15 Hz, with blocking of the nitrergic neurotransmitter pathway the slow wave increased from the no stimulation scenario in frequency by only 2.4x compared to 2.7x when the nitrergic pathway was not blocked. A 21 % reduction in slow wave amplitude occurred when the cholinergic pathway was blocked, compared to a 46% reduction when no neurotransmitter pathways were blocked. Clinical relevance - This mathematical model is a step towards successful computational modeling of the effects ther-apeutic neural stimulation on the stomach. The model is also a tool for understanding of the physiology of neural stimulation.
Collapse
|
40
|
Foong D, Liyanage L, Zhou J, Zarrouk A, Ho V, O'Connor MD. Single-cell RNA sequencing predicts motility networks in purified human gastric interstitial cells of Cajal. Neurogastroenterol Motil 2022; 34:e14303. [PMID: 34913225 DOI: 10.1111/nmo.14303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/25/2021] [Accepted: 11/17/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Gastrointestinal (GI) motility disorders affect millions of people worldwide, yet they remain poorly treated in part due to insufficient knowledge of the molecular networks controlling GI motility. Interstitial cells of Cajal (ICC) are critical GI pacemaker cells, and abnormalities in ICC are implicated in GI motility disorders. Two cell surface proteins, KIT and ANO1, are used for identifying ICC. However, difficulties accessing human tissue and the low frequency of ICC in GI tissues have meant human ICC are insufficiently characterized. Here, a range of characterization assays including single-cell RNA sequencing (scRNA-seq) was performed using KIT+ CD45- CD11B- primary human gastric ICC to better understand networks controlling human ICC biology. METHODS Excess sleeve gastrectomy tissues were dissected; ICC were analyzed by immunofluorescence, fluorescence-activated cell sorting (FACSorting), real-time PCR, mass spectrometry, and scRNA-seq. KEY RESULTS Immunofluorescence identified ANO1+ /KIT+ cells throughout the gastric muscle. Compared to the FACSorted negative cells, PCR showed the KIT+ CD45- CD11B- ICC were enriched 28-fold in ANO1 expression (p < 0.01). scRNA-seq analysis of the KIT- CD45+ CD11B+ and KIT+ CD45- CD11B- ICC revealed separate clusters of immune cells and ICC (respectively); cells in the ICC cluster expressed critical GI motility genes (eg, CAV1 and PRKG1). The scRNA-seq data for these two cell clusters predicted protein interaction networks consistent with immune cell and ICC biology, respectively. CONCLUSIONS & INFERENCES The single-cell transcriptome of purified KIT+ CD45- CD11B- human gastric ICC presented here provides new molecular insights and hypotheses into evolving models of GI motility. This knowledge will provide an improved framework to investigate targeted therapies for GI motility disorders.
Collapse
Affiliation(s)
- Daphne Foong
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Liwan Liyanage
- School of Computing, Data and Mathematical Sciences, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Jerry Zhou
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Ali Zarrouk
- Campbelltown Private Hospital, Campbelltown, New South Wales, Australia
| | - Vincent Ho
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia.,Campbelltown Private Hospital, Campbelltown, New South Wales, Australia
| | - Michael D O'Connor
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| |
Collapse
|
41
|
Integrating Network Pharmacology and In Vivo Model to Investigate the Mechanism of Biheimaer in the Treatment of Functional Dyspepsia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8773527. [PMID: 35668782 PMCID: PMC9166952 DOI: 10.1155/2022/8773527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022]
Abstract
Objective. Biheimaer (BHM) is a hospital formulation for clinical treatment of dyspepsia and acid reflux, based on Compatibility Theory of Traditional Chinese Medicine. This study anticipated to elucidate the molecular mechanism of BHM against Functional dyspepsia via combined network pharmacology prediction with experimental verification. Methods. Based on network pharmacology, the potential active components and targets of BHM in the treatment of functional dyspepsia were explored by prediction and molecular docking technology. The results of protein–protein interaction analysis, functional annotation, and pathway enrichment analysis further refined the main targets and pathways. The molecular mechanism of BHM improving functional dyspepsia mice induced by L-arginine + atropine was verified on the basis of network pharmacology. Results. In this study, 183 effective compounds were screened from BHM; moreover, 1007 compound-related predicted targets and 156 functional dyspepsia-related targets were found. The results of enrichment analysis and in vivo experiments showed that BHM could regulate intestinal smooth muscle contraction to play a therapeutic role in functional dyspepsia by reducing the expression of NOS3, SERT, TRPV1, and inhibiting the inflammatory cytokine (IL-1β, TNF-α) to intervene the inflammatory response in mice. Conclusions. This study revealed the molecular biological mechanisms of the Traditional Chinese Medicine formulation of BHM in functional dyspepsia by network pharmacology and experimental verification, meanwhile provided scientific support for subsequent clinical medication.
Collapse
|
42
|
Drumm BT, Cobine CA, Baker SA. Insights on gastrointestinal motility through the use of optogenetic sensors and actuators. J Physiol 2022; 600:3031-3052. [PMID: 35596741 DOI: 10.1113/jp281930] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/13/2022] [Indexed: 11/08/2022] Open
Abstract
The muscularis of the gastrointestinal (GI) tract consists of smooth muscle cells (SMCs) and various populations of interstitial cells of Cajal (ICC), platelet-derived growth factor receptor α+ (PDGFRα+ ) cells, as well as excitatory and inhibitory enteric motor nerves. SMCs, ICC and PDGFRα+ cells form an electrically coupled syncytium, which together with inputs from the enteric nervous system (ENS) regulate GI motility. Early studies evaluating Ca2+ signalling behaviours in the GI tract relied upon indiscriminate loading of tissues with Ca2+ dyes. These methods lacked the means to study activity in specific cells of interest without encountering contamination from other cells within the preparation. Development of mice expressing optogenetic sensors (GCaMP, RCaMP) has allowed visualization of Ca2+ signalling behaviours in a cell specific manner. Additionally, availability of mice expressing optogenetic modulators (channelrhodopsins or halorhodospins) has allowed manipulation of specific signalling pathways using light. GCaMP expressing animals have been used to characterize Ca2+ signalling behaviours of distinct classes of ICC and SMCs throughout the GI musculature. These findings illustrate how Ca2+ signalling in ICC is fundamental in GI muscles, contributing to tone in sphincters, pacemaker activity in rhythmic muscles and relaying enteric signals to SMCs. Animals that express channelrhodopsin in specific neuronal populations have been used to map neural circuitry and to examine post junctional neural effects on GI motility. Thus, optogenetic approaches provide a novel means to examine the contribution of specific cell types to the regulation of motility patterns within complex multi-cellular systems. Abstract Figure Legends Optogenetic activators and sensors can be used to investigate the complex multi-cellular nature of the gastrointestinal (GI tract). Optogenetic activators that are activated by light such as channelrhodopsins (ChR2), OptoXR and halorhodopsinss (HR) proteins can be genetically encoded into specific cell types. This can be used to directly activate or silence specific GI cells such as various classes of enteric neurons, smooth muscle cells (SMC) or interstitial cells, such as interstitial cells of Cajal (ICC). Optogenetic sensors that are activated by different wavelengths of light such as green calmodulin fusion protein (GCaMP) and red CaMP (RCaMP) make high resolution of sub-cellular Ca2+ signalling possible within intact tissues of specific cell types. These tools can provide unparalleled insight into mechanisms underlying GI motility and innervation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland.,Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Caroline A Cobine
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Salah A Baker
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
43
|
Koh SD, Drumm BT, Lu H, Kim HJ, Ryoo SB, Kim HU, Lee JY, Rhee PL, Wang Q, Gould TW, Heredia D, Perrino BA, Hwang SJ, Ward SM, Sanders KM. Propulsive colonic contractions are mediated by inhibition-driven poststimulus responses that originate in interstitial cells of Cajal. Proc Natl Acad Sci U S A 2022; 119:e2123020119. [PMID: 35446689 PMCID: PMC9170151 DOI: 10.1073/pnas.2123020119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/17/2022] [Indexed: 12/23/2022] Open
Abstract
The peristaltic reflex is a fundamental behavior of the gastrointestinal (GI) tract in which mucosal stimulation activates propulsive contractions. The reflex occurs by stimulation of intrinsic primary afferent neurons with cell bodies in the myenteric plexus and projections to the lamina propria, distribution of information by interneurons, and activation of muscle motor neurons. The current concept is that excitatory cholinergic motor neurons are activated proximal to and inhibitory neurons are activated distal to the stimulus site. We found that atropine reduced, but did not block, colonic migrating motor complexes (CMMCs) in mouse, monkey, and human colons, suggesting a mechanism other than one activated by cholinergic neurons is involved in the generation/propagation of CMMCs. CMMCs were activated after a period of nerve stimulation in colons of each species, suggesting that the propulsive contractions of CMMCs may be due to the poststimulus excitation that follows inhibitory neural responses. Blocking nitrergic neurotransmission inhibited poststimulus excitation in muscle strips and blocked CMMCs in intact colons. Our data demonstrate that poststimulus excitation is due to increased Ca2+ transients in colonic interstitial cells of Cajal (ICC) following cessation of nitrergic, cyclic guanosine monophosphate (cGMP)-dependent inhibitory responses. The increase in Ca2+ transients after nitrergic responses activates a Ca2+-activated Cl− conductance, encoded by Ano1, in ICC. Antagonists of ANO1 channels inhibit poststimulus depolarizations in colonic muscles and CMMCs in intact colons. The poststimulus excitatory responses in ICC are linked to cGMP-inhibited cyclic adenosine monophosphate (cAMP) phosphodiesterase 3a and cAMP-dependent effects. These data suggest alternative mechanisms for generation and propagation of CMMCs in the colon.
Collapse
Affiliation(s)
- Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Bernard T. Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Hongli Lu
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Hyun Jin Kim
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Seung-Bum Ryoo
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Heung-Up Kim
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Ji Yeon Lee
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Gangnam-Gu, Seoul, Korea 135-710
| | - Qianqian Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Thomas W. Gould
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Dante Heredia
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Brian A. Perrino
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| |
Collapse
|
44
|
Ability of 2-Chloro-N-(1-(3,4-dimethoxyphenyl)propan-2-yl)-2-phenylacetamide to Stimulate Endogenous Nitric Oxide Synthesis. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Papaverine is one of the isoquinoline alkaloids derived from opium which is a vasodilator and smooth muscle relaxant. Using its chemical structure as a basic model, we synthesized 2-chloro-N-(1-(3,4-dimethoxyphenyl)propan-2-yl)-2-phenylacetamide as an isoquinoline precursor (IQP). Aim: Clarifying the nature of the relationship between IQP as a new biologically active molecule and the neurotransmitters acetylcholine (ACh) and serotonin (5-hydroxytryptamine, 5-HT), as well as with the nitric oxide (NO). Materials and methods: The IQP compound was tested on the isolated gastric smooth muscle preparations (SMPs) from rats to determine its effects on spontaneous contractile activity. NO concentration in tissue homogenates was determined, and immunohistochemistry was used to visualize the expression of neuronal nitric oxide synthase (nNOS) and endothelial nitric oxide synthase (eNOS) in smooth muscle (SM) cells. Results: The data from the isometric measurements suggest that IQP has an additional specific action affecting the intracellular signaling pathways of 5-HT. Using immunohistochemistry, we found that the combination of 5-HT and IQP affected the density and intensity of nNOS-positive cells, which increase significantly in the myenteric plexus and SM cells. Conclusions: In conclusion, IQP is involved in the regulation of intestinal neurons expressing nNOS, affects the function of nNOS/NO, and, by this mechanism, probably regulates the spontaneous contractile activity of gastric SM.
Collapse
|
45
|
Chen Y, Zhang S, Li Y, Yan H, Ba Y, Wang X, Shi N, Liu C. Gastric Electrical Stimulation Increases the Proliferation of Interstitial Cells of Cajal and Alters the Enteric Nervous System in Diabetic Rats. Neuromodulation 2022; 25:1106-1114. [DOI: 10.1016/j.neurom.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/26/2022]
|
46
|
Forero-Rodríguez LJ, Josephs-Spaulding J, Flor S, Pinzón A, Kaleta C. Parkinson's Disease and the Metal-Microbiome-Gut-Brain Axis: A Systems Toxicology Approach. Antioxidants (Basel) 2021; 11:71. [PMID: 35052575 PMCID: PMC8773335 DOI: 10.3390/antiox11010071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/02/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's Disease (PD) is a neurodegenerative disease, leading to motor and non-motor complications. Autonomic alterations, including gastrointestinal symptoms, precede motor defects and act as early warning signs. Chronic exposure to dietary, environmental heavy metals impacts the gastrointestinal system and host-associated microbiome, eventually affecting the central nervous system. The correlation between dysbiosis and PD suggests a functional and bidirectional communication between the gut and the brain. The bioaccumulation of metals promotes stress mechanisms by increasing reactive oxygen species, likely altering the bidirectional gut-brain link. To better understand the differing molecular mechanisms underlying PD, integrative modeling approaches are necessary to connect multifactorial perturbations in this heterogeneous disorder. By exploring the effects of gut microbiota modulation on dietary heavy metal exposure in relation to PD onset, the modification of the host-associated microbiome to mitigate neurological stress may be a future treatment option against neurodegeneration through bioremediation. The progressive movement towards a systems toxicology framework for precision medicine can uncover molecular mechanisms underlying PD onset such as metal regulation and microbial community interactions by developing predictive models to better understand PD etiology to identify options for novel treatments and beyond. Several methodologies recently addressed the complexity of this interaction from different perspectives; however, to date, a comprehensive review of these approaches is still lacking. Therefore, our main aim through this manuscript is to fill this gap in the scientific literature by reviewing recently published papers to address the surrounding questions regarding the underlying molecular mechanisms between metals, microbiota, and the gut-brain-axis, as well as the regulation of this system to prevent neurodegeneration.
Collapse
Affiliation(s)
- Lady Johanna Forero-Rodríguez
- Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (L.J.F.-R.); (A.P.)
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Jonathan Josephs-Spaulding
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Stefano Flor
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Andrés Pinzón
- Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (L.J.F.-R.); (A.P.)
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| |
Collapse
|
47
|
Arterial blood flow waveform shapes – their original quantification and importance in chosen aspects of physiology and psychology: A review. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
48
|
Zhu GY, Jia DD, Yang Y, Miao Y, Wang C, Wang CM. The Effect of Shaoyao Gancao Decoction on Sphincter of Oddi Dysfunction in Hypercholesterolemic Rabbits via Protecting the Enteric Nervous System-Interstitial Cells of Cajal-Smooth Muscle Cells Network. J Inflamm Res 2021; 14:4615-4628. [PMID: 34552344 PMCID: PMC8450191 DOI: 10.2147/jir.s326416] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/17/2021] [Indexed: 11/25/2022] Open
Abstract
Objective This study observes the morphological changes in the enteric nervous system (ENS) – interstitial cells of Cajal (ICC) – smooth muscle cells (SMC) network in sphincter of Oddi dysfunction (SOD) in hypercholesterolemic rabbits following treatment with Shaoyao Gancao decoction (SGD), as well as the apoptosis of the ICC. Methods In this study, 48 healthy adult New Zealand rabbits are randomly divided into three groups (n = 16 in each group): the control, the model, and the SGD treatment groups. The hypercholesterolemic rabbit model is established. Hematoxylin and eosin staining, transmission electron microscopy, immunofluorescence, terminal deoxynucleotidyl transferase dUTP nick end labeling staining, immunohistochemistry, Western blot analysis, and reverse transcription-polymerase chain reaction are used to detect the morphological changes in the ENS–ICC–SMC network, the expression of apoptosis-related proteins in the ICC, and to observe the curative effect of SGD after treatment. Results Compared with the control group, the morphology and the ultrastructure of the SO are destroyed in the model group. In addition, the protein gene product 9.5 (PGP9.5), nitric oxide (NO), the SMCs, and the ICC all significantly decreased while substance P (SP) significantly increased. Compared with the model group, the SO morphology and ultrastructure are repaired in the SGD group. In addition, the PGP9.5, NO, the SMCs, and the ICC significantly increased while SP decreased. In addition, SGD may activate the stem cell factor (SCF)/c-Kit signaling pathway to treat SO dysfunction by up-regulating the expression of c-Kit and SCF. Similarly, this pathway restores SO by up-regulating the expression of Bcl2 and inhibiting cleaved caspase-3, Bax, and the tumor necrosis factor. Conclusion Shaoyao Gancao decoction can promote the recovery of sphincter of Oddi dysfunction in hypercholesterolemic rabbits by protecting the ENS–ICC–SMC network.
Collapse
Affiliation(s)
- Gui-Ying Zhu
- Department of General Surgery of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Dan-Dan Jia
- Department of General Surgery of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Ying Yang
- Department of General Surgery of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Ye Miao
- Department of General Surgery of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Chao Wang
- Department of General Surgery of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| | - Chang-Miao Wang
- Department of General Surgery of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, People's Republic of China
| |
Collapse
|
49
|
Nitric Oxide: From Gastric Motility to Gastric Dysmotility. Int J Mol Sci 2021; 22:ijms22189990. [PMID: 34576155 PMCID: PMC8470306 DOI: 10.3390/ijms22189990] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
It is known that nitric oxide (NO) plays a key physiological role in the control of gastrointestinal (GI) motor phenomena. In this respect, NO is considered as the main non-adrenergic, non-cholinergic (NANC) inhibitory neurotransmitter responsible for smooth muscle relaxation. Moreover, many substances (including hormones) have been reported to modulate NO production leading to changes in motor responses, further underlying the importance of this molecule in the control of GI motility. An impaired NO production/release has indeed been reported to be implicated in some GI dysmotility. In this article we wanted to focus on the influence of NO on gastric motility by summarizing knowledge regarding its role in both physiological and pathological conditions. The main role of NO on regulating gastric smooth muscle motor responses, with particular reference to NO synthases expression and signaling pathways, is discussed. A deeper knowledge of nitrergic mechanisms is important for a better understanding of their involvement in gastric pathophysiological conditions of hypo- or hyper-motility states and for future therapeutic approaches. A possible role of substances which, by interfering with NO production, could prove useful in managing such motor disorders has been advanced.
Collapse
|
50
|
Cairns BR, Jevans B, Chanpong A, Moulding D, McCann CJ. Automated computational analysis reveals structural changes in the enteric nervous system of nNOS deficient mice. Sci Rep 2021; 11:17189. [PMID: 34433854 PMCID: PMC8387485 DOI: 10.1038/s41598-021-96677-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
Neuronal nitric oxide synthase (nNOS) neurons play a fundamental role in inhibitory neurotransmission, within the enteric nervous system (ENS), and in the establishment of gut motility patterns. Clinically, loss or disruption of nNOS neurons has been shown in a range of enteric neuropathies. However, the effects of nNOS loss on the composition and structure of the ENS remain poorly understood. The aim of this study was to assess the structural and transcriptional consequences of loss of nNOS neurons within the murine ENS. Expression analysis demonstrated compensatory transcriptional upregulation of pan neuronal and inhibitory neuronal subtype targets within the Nos1-/- colon, compared to control C57BL/6J mice. Conventional confocal imaging; combined with novel machine learning approaches, and automated computational analysis, revealed increased interconnectivity within the Nos1-/- ENS, compared to age-matched control mice, with increases in network density, neural projections and neuronal branching. These findings provide the first direct evidence of structural and molecular remodelling of the ENS, upon loss of nNOS signalling. Further, we demonstrate the utility of machine learning approaches, and automated computational image analysis, in revealing previously undetected; yet potentially clinically relevant, changes in ENS structure which could provide improved understanding of pathological mechanisms across a host of enteric neuropathies.
Collapse
Affiliation(s)
- Ben R Cairns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N, UK
| | - Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N, UK
| | - Atchariya Chanpong
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N, UK
| | - Dale Moulding
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N, UK.
| |
Collapse
|