1
|
Desprez C, Leroi AM, Gourcerol G. Gastric and sacral electrical stimulation for motility disorders-A clinical perspective. Neurogastroenterol Motil 2024:e14884. [PMID: 39099155 DOI: 10.1111/nmo.14884] [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: 01/31/2024] [Revised: 06/04/2024] [Accepted: 07/21/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Electrical stimulation of the gut has been investigated in recent decades with a view to treating various gastro-intestinal motility disorders including, among others, gastric electrical stimulation to relieve nausea and vomiting associated with gastroparesis and sacral neuromodulation to treat fecal incontinence and/or constipation. Although their symptomatic efficacy has been ascertained by randomized controlled trials, their mechanisms of action are not fully understood. PURPOSE This review summarizes the past year's literature on the mechanisms of action of gut electrical stimulation therapies, including their impact on the gut-brain axis.
Collapse
Affiliation(s)
- Charlotte Desprez
- Digestive Physiology Department, Rouen University Hospital, Rouen, France
| | - Anne-Marie Leroi
- Digestive Physiology Department, Rouen University Hospital, Rouen, France
| | | |
Collapse
|
2
|
Lv L, Li W, Guo D, Shi B, Li Y. Early Sacral Neuromodulation Prevented Detrusor Overactivity in Rats With Spinal Cord Injury. Neuromodulation 2024:S1094-7159(24)00629-9. [PMID: 39046393 DOI: 10.1016/j.neurom.2024.05.008] [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: 03/12/2024] [Revised: 05/05/2024] [Accepted: 05/24/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVES Sacral neuromodulation (SNM) has been shown to alleviate bladder dysfunction in patients with overactive bladder and nonobstructive urinary retention. However, the therapeutic effect and mechanism of SNM in neurogenic bladder dysfunction are still not fully understood. Using a rat model of spinal cord injury (SCI), this study aims to investigate the therapeutic effect of early SNM in the bladder-areflexia phase on neurogenic bladder dysfunction and evaluate its possible mechanism. MATERIALS AND METHODS Basic physiological parameters such as body/bladder weight, blood pressure, and electrocardiogram results were measured to evaluate the safety of SNM. Enzyme-linked immunosorbent assays and quantitative real-time polymerase chain reaction were used to examine the expression of proinflammatory factors. Hematoxylin and eosin and Masson's trichrome staining were used to observe morphological changes, and cystometry was used to evaluate urodynamic changes after SNM treatment. Western blotting and immunofluorescence staining were used to measure the levels of transient receptor potential vanilloid 1 (TRPV1) and calcitonin gene-related peptide (CGRP) in the L6-S1 dorsal root ganglia (DRGs) and bladder. Capsaicin desensitization was used to investigate whether inhibiting TRPV1 could prevent detrusor overactivity in SCI rats. RESULTS Early SNM did not affect the body/bladder weight, heart rate, blood pressure, or the expression of proinflammatory cytokines (PGE2, IL-1, IL-2, IL-6, TGF-β, or TNF-α) in the bladders of SCI rats. Morphologically, early SNM prevented urothelial edema (p = 0.0248) but did not influence collagen/smooth muscle in the bladder. Compared with untreated rats with SCI, the rats treated with SNM exhibited increased bladder capacity (p = 0.0132) and voiding efficiency (p = 0.0179), and decreased nonvoiding contraction (NVC) frequency (p = 0.0240). The maximum pressure, basal pressure, postvoid residual, and NVC amplitude did not change significantly. After the SNM treatment, the expression of TRPV1 in the bladder and CGRP in L6-S1 DRGs weredecreased (L6, p = 0.0160; S1, p = 0.0024) in SCI rats. In capsaicin-desensitized SCI rats, urodynamic results showed an increase in bladder capacity (p = 0.0116) and voiding efficiency (p = 0.0048), and diminished NVC frequency (p = 0.0116), while other parameters did not change significantly. CONCLUSIONS Early SNM prevented urothelial edema morphologically and detrusor overactivity in SCI rats. Inhibition of TRPV1 in the bladder and DRGs may be one of the potential mechanisms for preventing detrusor overactivity by SNM.
Collapse
Affiliation(s)
- Linchen Lv
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China; University of Health and Rehabilitation Sciences, Qingdao, China
| | - Wenxian Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China; Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dongyue Guo
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China; University of Health and Rehabilitation Sciences, Qingdao, China.
| | - Yan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Urinary Precision Diagnosis and Treatment in Universities of Shandong, Jinan, China.
| |
Collapse
|
3
|
McKay DM, Defaye M, Rajeev S, MacNaughton WK, Nasser Y, Sharkey KA. Neuroimmunophysiology of the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2024; 326:G712-G725. [PMID: 38626403 PMCID: PMC11376980 DOI: 10.1152/ajpgi.00075.2024] [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: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/18/2024]
Abstract
Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.
Collapse
Affiliation(s)
- Derek M McKay
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sruthi Rajeev
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
4
|
Roelandt P, Bislenghi G, Coremans G, De Looze D, Denis MA, De Schepper H, Dewint P, Geldof J, Gijsen I, Komen N, Ruymbeke H, Stijns J, Surmont M, Van de Putte D, Van den Broeck S, Van Geluwe B, Wyndaele J. Belgian consensus guideline on the management of anal fissures. Acta Gastroenterol Belg 2024; 87:304-321. [PMID: 39210763 DOI: 10.51821/87.2.11787] [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] [Indexed: 09/04/2024]
Abstract
Introduction Acute and chronic anal fissures are common proctological problems that lead to relatively high morbidity and frequent contacts with health care professionals. Multiple treatment options, both topical and surgical, are available, therefore evidence-based guidance is preferred. Methods A Delphi consensus process was used to review the literature and create relevant statements on the treatment of anal fissures. These statements were discussed and modulated until sufficient agreement was reached. These guidelines were based on the published literature up to January 2023. Results Anal fissures occur equally in both sexes, mostly between the second and fourth decades of life. Diagnosis can be made based on cardinal symptoms and clinical examination. In case of insufficient relief with conservative treatment options, pharmacological sphincter relaxation is preferred. After 6-8 weeks of topical treatment, surgical options can be explored. Both lateral internal sphincterotomy as well as fissurectomy are well-established surgical techniques, both with specific benefits and risks. Conclusions The current guidelines for the management of anal fissures include recommendations for the clinical evaluation of anal fissures, and their conservative, topical and surgical management.
Collapse
Affiliation(s)
- P Roelandt
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
- Translational Research in Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - G Bislenghi
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - G Coremans
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - D De Looze
- Department of Gastroenterology and Hepatology, University Hospital Ghent, Ghent, Belgium
| | - M A Denis
- Department of Gastroenterology and Hepatology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - H De Schepper
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - P Dewint
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
- Department of Gastroenterology and Hepatology, Maria Middelares Hospital, Ghent, Belgium
| | - J Geldof
- Department of Gastroenterology and Hepatology, University Hospital Ghent, Ghent, Belgium
| | - I Gijsen
- Department of Gastroenterology and Hepatology, Noorderhart Hospital, Pelt, Belgium
| | - N Komen
- Department of Abdominal Surgery, Antwerp University Hospital, Edegem, Belgium
- Antwerp RESURG Group, Antwerp Surgical Training, Anatomy and Research Centre (ASTARC), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - H Ruymbeke
- Department of Gastroenterology and Hepatology, University Hospital Ghent, Ghent, Belgium
- Department of Gastroenterology, VITAZ, Sint-Niklaas, Belgium
| | - J Stijns
- Department of Abdominal Surgery, University Hospital Brussels, Brussels, Belgium
| | - M Surmont
- Department of Gastroenterology and Hepatology, University Hospital Brussels, Brussels, Belgium
| | - D Van de Putte
- Department of Gastro-intestinal Surgery, University Hospital Ghent, Ghent, Belgium
| | - S Van den Broeck
- Department of Abdominal Surgery, Antwerp University Hospital, Edegem, Belgium
| | - B Van Geluwe
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Abdominal Surgery, General Hospital Groeninge, Kortrijk, Belgium
| | - J Wyndaele
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Ocampo YC, Castro JP, Pájaro IB, Caro D, Talero E, Motilva V, Franco LA. Protective effect of sucrose esters from cape gooseberry (Physalis peruviana L.) in TNBS-induced colitis. PLoS One 2024; 19:e0299687. [PMID: 38512973 PMCID: PMC10957089 DOI: 10.1371/journal.pone.0299687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/15/2024] [Indexed: 03/23/2024] Open
Abstract
Phytotherapy is an attractive strategy to treat inflammatory bowel disease (IBD) that could be especially useful in developing countries. We previously demonstrated the intestinal anti-inflammatory effect of the total ethereal extract from the Physalis peruviana (Cape gooseberry) calyces in TNBS-induced colitis. This work investigates the therapeutic potential of Peruviose A and B, two sucrose esters that constitute the major metabolites of its calyces. The effect of the Peruvioses A and B mixture on TNBS-induced colitis was studied after 3 (preventive) and 15-days (therapy set-up) of colitis induction in rats. Colonic inflammation was assessed by measuring macroscopic/histologic damage, MPO activity, and biochemical changes. Additionally, LPS-stimulated RAW 264.7 macrophages were treated with test compounds to determine the effect on cytokine imbalance in these cells. Peruvioses mixture ameliorated TNBS-induced colitis in acute (preventive) or established (therapeutic) settings. Although 3-day treatment with compounds did not produce a potent effect, it was sufficient to significantly reduce the extent/severity of tissue damage and the microscopic disturbances. Beneficial effects in the therapy set-up were substantially higher and involved the inhibition of pro-inflammatory enzymes (iNOS, COX-2), cytokines (TNF-α, IL-1β, and IL-6), as well as epithelial regeneration with restoration of goblet cells numbers and expression of MUC-2 and TFF-3. Consistently, LPS-induced RAW 264.7 cells produced less NO, PGE2, TNF-α, IL-6, and MCP-1. These effects might be related to the inhibition of the NF-κB signaling pathway. Our results suggest that sucrose esters from P. peruviana calyces, non-edible waste from fruit production, might be useful as an alternative IBD treatment.
Collapse
Affiliation(s)
- Yanet C. Ocampo
- Biological Evaluation of Promising Substances Group, Faculty of Pharmaceutical Science, Universidad de Cartagena, Cartagena, Colombia
| | - Jenny P. Castro
- Biological Evaluation of Promising Substances Group, Faculty of Pharmaceutical Science, Universidad de Cartagena, Cartagena, Colombia
- Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla, Colombia
| | - Indira B. Pájaro
- Biological Evaluation of Promising Substances Group, Faculty of Pharmaceutical Science, Universidad de Cartagena, Cartagena, Colombia
- Faculty of Chemistry and Pharmacy, Universidad del Atlántico, Barranquilla, Colombia
| | - Daneiva Caro
- Biological Evaluation of Promising Substances Group, Faculty of Pharmaceutical Science, Universidad de Cartagena, Cartagena, Colombia
| | - Elena Talero
- Department of Pharmacology, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Virginia Motilva
- Department of Pharmacology, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Luis A. Franco
- Biological Evaluation of Promising Substances Group, Faculty of Pharmaceutical Science, Universidad de Cartagena, Cartagena, Colombia
| |
Collapse
|
6
|
Zhang S, Zhang C, Yan H, Yang L, Shi N, Liu C, Chen Y. Sacral Nerve Stimulation Alleviates Intestinal Inflammation Through Regulating the Autophagy of Macrophages and Activating the Inflammasome Mediated by a Cholinergic Antiinflammatory Pathway in Colitis Rats. Neuromodulation 2024; 27:302-311. [PMID: 36740464 DOI: 10.1016/j.neurom.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/16/2022] [Accepted: 01/07/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized by chronic progressive intestinal inflammation. Sacral nerve stimulation (SNS) ameliorates colon inflammation caused by IBD. The aim of this study was to investigate the antiinflammatory benefits of SNS in colitis rats and explore the roles of the cholinergic antiinflammatory pathway, macrophage autophagy, and nucleotide oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3) inflammatory bodies. MATERIALS AND METHODS Rats were divided into four groups: healthy control, dextran sulfate sodium (DSS), DSS + sham-SNS, and DSS + SNS groups. An electrode was surgically placed in the right sacral nerve (S3) for stimulation. The disease activity index (DAI) score was recorded each day, and the degree of inflammatory injury was evaluated using hematoxylin and eosin staining. The alpha7 nicotinic acetylcholine receptor (α7nAChR) and autophagy- and NLRP3-related factors were assessed using immunofluorescence staining and Western blotting. RESULTS The DSS group showed a higher DAI score, colon shortening, upregulated proinflammatory action, and colon damage, and the DSS + SNS group showed significantly improved symptoms. The number of α7nAChR+ cells and the expression level of autophagy decreased in the DSS group but increased in the DSS + SNS group. Conversely, the DSS group showed increased activation of NLRP3 inflammatory bodies, whereas the DSS + SNS group showed decreased activation of NLRP3 inflammatory bodies. CONCLUSION In this study, SNS ameliorated colon inflammation by enhancing macrophage autophagy and inhibiting the activation of NLRP3 inflammatory bodies, which may be related to the opening of the cholinergic antiinflammatory pathway.
Collapse
Affiliation(s)
- Shuhui Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Can Zhang
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Hui Yan
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lijuan Yang
- Cancer Research Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ning Shi
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chengxia Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yan Chen
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
7
|
Chen Z, Li J, Ma Q, Pikov V, Li M, Wang L, Liu Y, Ni M. Anti-Inflammatory Effects of Two-Week Sacral Nerve Stimulation Therapy in Patients With Ulcerative Colitis. Neuromodulation 2024; 27:360-371. [PMID: 37055336 DOI: 10.1016/j.neurom.2023.01.019] [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: 02/11/2022] [Revised: 12/24/2022] [Accepted: 01/03/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND AND AIMS Sacral nerve stimulation (SNS) showed anti-inflammatory properties in animal models of inflammatory bowel disease. We aimed to evaluate the effectiveness and safety of SNS in patients with ulcerative colitis (UC). MATERIALS AND METHODS Twenty-six patients with mild and moderate disease were randomized into two groups: SNS (delivered at S3 and S4 sacral foramina) and sham-SNS (delivered 8-10 mm away from sacral foramina), with the therapy applied once daily for one hour, for two weeks. We evaluated the Mayo score and several exploratory biomarkers, including C-reactive protein in the plasma, pro-inflammatory cytokines and norepinephrine in the serum, assessment of autonomic activity, and diversity and abundance of fecal microbiota species. RESULTS After two weeks, 73% of the subjects in the SNS group achieved clinical response, compared with 27% in the sham-SNS group. Levels of C-reactive protein, pro-inflammatory cytokines in the serum, and autonomic activity were significantly improved toward a healthy profile in the SNS group but not in the sham-SNS group. Absolute abundance of fecal microbiota species and one of the metabolic pathways were changed in the SNS group but not in the sham-SNS group. Significant correlations were observed between pro-inflammatory cytokines and norepinephrine in the serum on the one side and fecal microbiota phyla on the other side. CONCLUSIONS Patients with mild and moderate UC were responsive to a two-week SNS therapy. After performing further studies to evaluate its efficacy and safety, temporary SNS delivered through acupuncture needles may become a useful screening tool for identifying SNS therapy responders before considering long-term implantation of the implantable pulse generator and SNS leads for performing long-term SNS therapy.
Collapse
Affiliation(s)
- Zhengxin Chen
- National Center for Colorectal Diseases, Nanjing Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Jing Li
- Department of Acupuncture and Moxibustion, Nanjing Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Qiyao Ma
- Graduate School, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China; Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, Jiangsu Province, China
| | | | - Min Li
- National Center for Colorectal Diseases, Nanjing Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Ling Wang
- Graduate School, Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Ying Liu
- National Center for Colorectal Diseases, Nanjing Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Min Ni
- National Center for Colorectal Diseases, Nanjing Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China.
| |
Collapse
|
8
|
Pikov V. Vagus Nerve Stimulation and Sacral Nerve Stimulation for Inflammatory Bowel Disease: A Systematic Review. JOURNAL OF TRANSLATIONAL GASTROENTEROLOGY 2023; 1:94-100. [PMID: 38606364 PMCID: PMC11007757 DOI: 10.14218/jtg.2023.00098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2024]
Abstract
Background and objectives In this systematic review, we assessed the efficacy, potential mechanisms, and safety of two neuromodulation therapies in patients with inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis. The first therapy is vagus nerve stimulation (VNS) utilizing implantable or transcutaneous electrodes, and the second is sacral nerve stimulation (SNS) using implantable or percutaneous electrodes. Methods We conducted a systematic literature review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The PubMed database was comprehensively searched, and studies were rigorously assessed for inclusion and exclusion criteria. Results Our analysis encompassed five clinical studies, three on VNS and two on SNS. Most investigated studies demonstrated significant beneficial effects on IBD symptoms, including disease activity, severity of intestinal lesions, and intestinal pain. When evaluating the impact on key IBD pathophysiologies, both VNS and SNS exhibited trends toward reducing biomarkers of intestinal mucosal inflammation and mitigating sympathetic dominance. Importantly, none of the evaluated neuromodulation methods resulted in long-term adverse effects. Conclusions Cumulative evidence from the evaluated studies indicates that VNS and SNS therapies effectively alleviate IBD symptoms and may hold promise in addressing the underlying pathophysiologies of IBD, including intestinal mucosal inflammation and sympathetic dominance. Consequently, they represent valuable options for individualized IBD treatment.
Collapse
|
9
|
Wang X, Chen JDZ. Therapeutic potential and mechanisms of sacral nerve stimulation for gastrointestinal diseases. J Transl Int Med 2023; 11:115-127. [PMID: 37408571 PMCID: PMC10318922 DOI: 10.2478/jtim-2023-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
Background The aim of this systemtic review is to introduce clinical applications (especially emerging) and potential mechanisms of sacral nerve stimulation (SNS) for treating various gastrointestinal diseases. Materials and Methods PubMed and Web of Science were searched for studies published on SNS and its clinical applications in fecal incontinence (limited to systematic review and meta-analysis of clinical studies), constipation (limited to reviews and randomized control clinical studies), irritable bowel syndrome (IBS), inflammatory bowel disease (IBD) and upper gastrointestinal motility disorders. The relevant studies were pooled, and their findings were summarized and discussed. Results SNS is an approved method for treating fecal incontinence. Systematic review and meta-analysis demonstrated high efficacy of the SNS therapy for fecal incontinence. Increased anal sphincter pressure and improvement in rectal sensation were reported as major mechanisms involved in the SNS therapy. SNS has also been proposed for treating constipation, but the therapy has been shown ineffective. There is a lack in SNS methodological optimization and mechanistic research. A few basic and clinical studies have reported the potential of SNS for treating visceral pain in IBS. SNS seemed capable of improving mucosal barrier functions. Several case reports are available in the literature on the treatment of IBD with SNS. Several laboratory studies suggested therapeutic potential of a special method of SNS for IBD. Cholinergic anti-inflammatory mechanisms were reported. Due to a recently reported spinal afferent and vagal efferent pathway of SNS, a few preclinical studies reported the potential of SNS for upper gastrointestinal motility disorders. However, no clinical studies have been performed. Conclusions SNS for fecal incontinence is a well-established clinical therapy. However, the current method of SNS is ineffective for treating constipation. Further methodological development and randomized clinical trials are needed to explore potential applications of SNS for IBS and IBD.
Collapse
Affiliation(s)
- Ximeng Wang
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD21228, USA
| | - Jiande DZ Chen
- Department of Internal Medicine, University of Michigan School of Medicine, Ann ArborMI48109, USA
| |
Collapse
|
10
|
Drissi F, Bourreille A, Neunlist M, Meurette G. Sacral neuromodulation for refractory ulcerative colitis: safety and efficacy in a prospective observational series of eight patients. Tech Coloproctol 2023; 27:501-505. [PMID: 37043102 PMCID: PMC10169876 DOI: 10.1007/s10151-023-02793-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/16/2023] [Indexed: 04/13/2023]
Abstract
PURPOSE Ulcerative colitis (UC) treatment is mainly based on immunosuppressive therapy. As anti-inflammatory effects of sacral neuromodulation (SNM) have been previously reported in animal models, we conducted a pilot study aimed at assessing clinical, biological, and endoscopic response but also safety of SNM use in UC refractory to medical therapy. METHODS Adult patients with histologically proven UC resistant to immunosuppressive therapy were invited to enroll in the study. Primary outcome was the rate of UC remission (UCDAI score ≤ 2, without any criteria > 1) at 8 weeks (W8). Secondary outcomes were biological and endoscopic response also evaluated at W8 and W16. Subsequently, every patient was followed every 6 months. Adverse events were prospectively collected for safety assessment during the follow-up. RESULTS Eight patients, with mean age 47 years old, suffering from UC for 2-13 years were included. There were no complications in relation to SNM procedure. The acceptance of the device was excellent in all patients. Clinical and endoscopic remission was obtained at W8 in one patient (12.5%) and three other patients (37.5%) were responders at W16. At review (mean follow-up of 4 years), two patients (25%) were in remission and two (25%) were responders. CONCLUSION SNM application is safe in patients suffering from refractory UC. Effects on disease activity were mainly observed after 16 weeks. Larger prospective studies are mandatory, but SNM could be a way to reinforce medical therapy and reduce the use of immunosuppressive drugs.
Collapse
Affiliation(s)
- Farouk Drissi
- Chirurgie Cancérologique Digestive et Endocrinienne, Institut des Maladies de l'Appareil Digestif, University Hospital of Nantes, Hôtel Dieu, 1 Place Alexis Ricordeau, 44093, Nantes, Cedex 01, France
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l'Appareil Digestif, University of Nantes, Inserm, TENS, Nantes, France
- University Hospital of Nantes, Hôtel Dieu, Nantes, France
| | - Arnaud Bourreille
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l'Appareil Digestif, University of Nantes, Inserm, TENS, Nantes, France
- University Hospital of Nantes, Hôtel Dieu, Nantes, France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l'Appareil Digestif, University of Nantes, Inserm, TENS, Nantes, France
- University Hospital of Nantes, Hôtel Dieu, Nantes, France
| | - Guillaume Meurette
- The Enteric Nervous System in Gut and Brain Disorders, Institut des Maladies de l'Appareil Digestif, University of Nantes, Inserm, TENS, Nantes, France.
- Division of Digestive Surgery, University Hospital of Geneva, Geneva, Switzerland.
| |
Collapse
|
11
|
Pikov V. Bioelectronic medicine for restoring autonomic balance in autoimmune diseases. GUT MICROBIOTA AND INTEGRATIVE WELLNESS 2023; 1:182. [PMID: 37155473 PMCID: PMC10125261 DOI: 10.54844/gmiw.2022.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The aim of this mini-review is to introduce most prevalent autoimmune diseases, emphasize the importance of sympatho-parasympathetic imbalance in these autoimmune diseases, demonstrate how such imbalance can be effectively treated using the bioelectronic medicine, and describe potential mechanisms of bioelectronic medicine effects on the autoimmune activity at the cellular and molecular levels.
Collapse
|
12
|
Yasmin F, Sahito AM, Mir SL, Khatri G, Shaikh S, Gul A, Hassan SA, Koritala T, Surani S. Electrical neuromodulation therapy for inflammatory bowel disease. World J Gastrointest Pathophysiol 2022; 13:128-142. [PMID: 36187600 PMCID: PMC9516456 DOI: 10.4291/wjgp.v13.i5.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/19/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease of the gastrointestinal (GI) tract. It has financial and quality of life impact on patients. Although there has been a significant advancement in treatments, a considerable number of patients do not respond to it or have severe side effects. Therapeutic approaches such as electrical neuromodulation are being investigated to provide alternate options. Although bioelectric neuromodulation technology has evolved significantly in the last decade, sacral nerve stimulation (SNS) for fecal incontinence remains the only neuromodulation protocol commonly utilized use for GI disease. For IBD treatment, several electrical neuromodulation techniques have been studied, such as vagus NS, SNS, and tibial NS. Several animal and clinical experiments were conducted to study the effectiveness, with encouraging results. The precise underlying mechanisms of action for electrical neuromodulation are unclear, but this modality appears to be promising. Randomized control trials are required to investigate the efficacy of intrinsic processes. In this review, we will discuss the electrical modulation therapy for the IBD and the data pertaining to it.
Collapse
Affiliation(s)
- Farah Yasmin
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Abdul Moiz Sahito
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Syeda Lamiya Mir
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Govinda Khatri
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Somina Shaikh
- Department of Medicine, Dow University of Health Sciences, Karachi 74200, Pakistan
| | - Ambresha Gul
- Department of Medicine, People’s University of Medical and Health Sciences, Nawabshah 67480, Pakistan
| | - Syed Adeel Hassan
- Department of Medicine, University of Louisville, Louiseville, KY 40292, United States
| | - Thoyaja Koritala
- Department of Medicine, Mayo Clinic, Rochester, NY 55902, United States
| | - Salim Surani
- Department of Medicine, Texas A&M University, College Station, TX 77843, United States
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55902, United States
| |
Collapse
|
13
|
Perivoliotis K, Baloyiannis I, Ragias D, Beis N, Papageorgouli D, Xydias E, Tepetes K. The role of percutaneous tibial nerve stimulation (PTNS) in the treatment of chronic anal fissure: a systematic review. Int J Colorectal Dis 2021; 36:2337-2346. [PMID: 34132862 DOI: 10.1007/s00384-021-03976-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE This study was designed to summarize the current evidence regarding the role of percutaneous tibial nerve stimulation (PTNS) in the treatment of chronic anal fissure (CAF). METHODS The present systematic review of the literature was conducted on the basis of the PRISMA guidelines and the Cochrane Handbook for Systematic Reviews of Interventions. The primary endpoint of our study was the CAF recurrence rate. Quality assessment was based on the RoB 2 tool and the Case Series Quality Checklist. RESULTS Overall, 5 studies and 102 patients were included. A considerably heterogeneity in the neuromodulation technique and setting was identified. The pooled recurrence rate was estimated at the level of 19% (16/84). Post-interventional pain and Wexner scores were considerably reduced. The 2-month healing rate was 72% (18/25), whereas 73.6% of patients were symptom-free at 6 months. CONCLUSIONS PTNS is an effective alternative for the non-operative management of CAF. Due to several limitations further larger and higher quality studies are required.
Collapse
Affiliation(s)
| | - Ioannis Baloyiannis
- Department of Surgery, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| | - Dimitrios Ragias
- Faculty of Medicine, University of Thessaly, Mezourlo, 41110, Larissa, Greece
| | - Nikolaos Beis
- Faculty of Medicine, University of Thessaly, Mezourlo, 41110, Larissa, Greece
| | | | - Emmanouil Xydias
- Faculty of Medicine, University of Thessaly, Mezourlo, 41110, Larissa, Greece
| | - Konstantinos Tepetes
- Department of Surgery, University Hospital of Larissa, Mezourlo, 41110, Larissa, Greece
| |
Collapse
|
14
|
Abstract
INTRODUCTION Sacral neuromodulation (SNM) aims to improve anorectal function in patients with disorders of anal continence and rectal emptying. The mechanism of action of SNM is not well known, and its indications are still under evaluation. We report the functional results and morbidity of a prospective cohort treated between 2002 and 2019. RESULTS A total of 284 patients (of 423 tested) had implantation of a SNM. Five patients (1.8%) were lost to follow-up. Among those who had implantation, the indications for SNM were anal incontinence (n=376), refractory constipation (n=17), anterior resection syndrome (n=13), solitary rectal ulcer syndrome (n=7), and chronic inflammatory bowel disease (IBD) (n=10). The morbidity rate was 2.7% (Dindo-Clavien>2), 33 patients (11%) required explantation for infection (n=5), pain (n=2), inefficacy (n=24) or other reasons (rectal cancer) (n=3). It was necessary to change the stimulator in 68 patients (24%) during the follow-up period. Regarding the group of patients with anal incontinence, functional results showed improvement of the incontinence score in 40% and of quality of life in 25% after a mean follow-up of 55months. CONCLUSION SNM constitutes a mini-invasive treatment associated with low morbidity. Its' efficacy in anal incontinence makes it a priority approach. Other indications are still under evaluation; while results are promising, they are highly variable.
Collapse
|
15
|
Polysaccharides in natural products that repair the damage to intestinal mucosa caused by cyclophosphamide and their mechanisms: A review. Carbohydr Polym 2021; 261:117876. [PMID: 33766363 DOI: 10.1016/j.carbpol.2021.117876] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 12/18/2022]
Abstract
Cyclophosphamide (CTX) is a commonly used antitumor drug in clinical practice, and intestinal mucosal injury is one of its main toxic side effects, which seriously affects the treatment tolerance and prognosis of patients. Therefore, the prevention of intestinal mucosal injury is a research hotspot. Studies have shown that polysaccharides can effectively prevent and improve CTX-induced intestinal mucosal injury and immune system disorders. Recent research has elucidated the structure, biological function, and physicochemical properties of polysaccharides that prevent intestinal mucosal injury, and the potential mechanisms whereby they have this effect. In this paper, we review the recent progress made in understanding the effects of polysaccharides on intestinal mucosal injury and their protective mechanism in order to provide a reference for further research on the prevention of intestinal mucosal injury and the mechanisms involved in nutritional intervention.
Collapse
|
16
|
Tu L, Gharibani P, Yin J, Chen JDZ. Sacral nerve stimulation ameliorates colonic barrier functions in a rodent model of colitis. Neurogastroenterol Motil 2020; 32:e13916. [PMID: 32537873 DOI: 10.1111/nmo.13916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 03/30/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The mucosal barrier damage is recognized as one of the key factors in the pathogenesis of colitis. While sacral nerve stimulation (SNS) was reported to have therapeutic potential for colitis, its mechanisms of actions on colonic permeability remained largely unknown. METHODS In this study, colitis was induced by intrarectal administration of TNBS in rats. Five days later, they were treated with SNS or sham-SNS for 10 days. The effects of SNS on colonic permeability were assessed by measuring the expression of tight-junction proteins involved in regulating permeability and the FITC-dextran test. The mechanism of actions of SNS was investigated by studying the function of the enteric nervous system (ENS) cells and analyzing the autonomic nervous system. KEY RESULTS SNS decreased the disease activity index, microscopic and macroscopic scores, myeloperoxidase activity, and pro-inflammatory cytokines (TNF-α, IL-6). SNS increased the expression of Zonula Occludens-1, Occludin, Claudin-1, and Junctional adhesion molecule-A in the colon tissue. The FITC-dextran test showed that the colonic permeability was lower with SCS than sham-SNS. SNS increased ChAT, pancreatic polypeptide, and GDNF and reduced norepinephrine NGF, sub-P, and mast cell overactivation in the colon tissue. Concurrently, SNS increased acetylcholine in colon tissues and elevated vagal efferent activity. CONCLUSIONS & INFERENCES SNS ameliorates colonic inflammation and enhances colonic barrier function with the proposed mechanisms involving the increase in parasympathetic activity and modulation of the activity of the ENS and immune system, including mast cells.
Collapse
Affiliation(s)
- Lei Tu
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Payam Gharibani
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jieyun Yin
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiande D Z Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Cheng J, Shen H, Chowdhury R, Abdi T, Selaru F, Chen JDZ. Potential of Electrical Neuromodulation for Inflammatory Bowel Disease. Inflamm Bowel Dis 2020; 26:1119-1130. [PMID: 31782957 DOI: 10.1093/ibd/izz289] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD) is a common chronic inflammatory disease of the digestive tract that is often debilitating. It affects patients' quality of life and imposes a financial burden. Despite advances in treatment with medications such as biologics, a large proportion of patients do not respond to medical therapy or develop adverse events. Therefore, alternative treatment options such as electrical neuromodulation are currently being investigated. Electrical neuromodulation, also called bioelectronic medicine, is emerging as a potential new treatment for IBD. Over the past decade, advancements have been made in electrical neuromodulation. A number of electrical neuromodulation methods, such as vagus nerve stimulation, sacral nerve stimulation, and tibial nerve stimulation, have been tested to treat IBD. A series of animal and clinical trials have been performed to evaluate efficacy with promising results. Although the exact underlying mechanisms of action for electrical neuromodulation remain to be explored, this modality is promising. Further randomized controlled trials and basic experiments are needed to investigate efficacy and clarify intrinsic mechanisms.
Collapse
Affiliation(s)
- Jiafei Cheng
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Division of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Shen
- Division of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Reezwana Chowdhury
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tsion Abdi
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Florin Selaru
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiande D Z Chen
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Tu L, Gharibani P, Zhang N, Yin J, Chen JD. Anti-inflammatory effects of sacral nerve stimulation: a novel spinal afferent and vagal efferent pathway. Am J Physiol Gastrointest Liver Physiol 2020; 318:G624-G634. [PMID: 32068444 DOI: 10.1152/ajpgi.00330.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sacral nerve stimulation (SNS) was reported to improve 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis in rats. The aim of this study was to investigate whether the SNS anti-inflammatory effect is mediated via the local sacral splanchnic nerve or the spinal afferent-vagal efferent-colon pathway. Under general anesthesia, rats were administrated with TNBS intrarectally, and bipolar SNS electrodes were implanted unilaterally at S3. The sacral and vagal nerves were severed at different locations for the assessment of the neural pathway. SNS for 10 days improved colonic inflammation only in groups with intact afferent sacral nerve and vagus efferent nerve. SNS markedly increased acetylcholine and anti-inflammatory cytokines (IL-10) and decreased myeloperoxidase and proinflammatory cytokines (IL-2, IL-17A, and TNF-α) in colon tissues. SNS increased the number of c-fos-positive cells in the brain stem and normalized vagal activity measured by spectral analysis of heart rate variability. SNS exerts an anti-inflammatory effect on TNBS-induced colitis by enhancing vagal activity mediated mainly via the spinal afferent-brain stem-vagal efferent-colon pathway.NEW & NOTEWORTHY Our findings support that there is a possible sacral afferent-vagal efferent pathway that can transmit sacral nerve stimulation to the colon tissue. Sacral nerve stimulation can be carried out by spinal cord afferent to the brain stem and then by the vagal nerve (efferent) to the target organ.
Collapse
Affiliation(s)
- Lei Tu
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Payam Gharibani
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nina Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jieyun Yin
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jiande Dz Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Pasricha TS, Zhang H, Zhang N, Chen JDZ. Sacral nerve stimulation prompts vagally-mediated amelioration of rodent colitis. Physiol Rep 2020; 8:e14294. [PMID: 31925899 PMCID: PMC6954119 DOI: 10.14814/phy2.14294] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022] Open
Abstract
Neuromodulation based on the vagal anti-inflammatory reflex has emerged as an exciting therapeutic approach for chronic inflammatory diseases. However, it is unclear whether direct stimulation of the vagus or of pelvic nerves coming from sacral roots, providing the bulk of colonic parasympathetic innervation, is the best approach. We hypothesized that sacral nerve stimulation (SNS) would be an effective treatment for colitis. Age and sex-matched Sprague-Dawley rats were administered 5% dextran sulphate sodium (DSS) in drinking water ad libitum for 7 days. A group of rats was sacrificed after DSS treatment, and the remaining rats were randomized to either sham-SNS or SNS groups, which were performed for 1 hr daily for 10 days. Stimulations were delivered via chronically implanted electrodes using an 8-channel universal pulse generator. Sacral nerve stimulation promoted recovery of colitis demonstrated by decreased disease activity index, myeloperoxidase activity, tissue TNF-alpha, and histological scores as well as an increased colonic M2 macrophage population. Heart rate variability analysis demonstrated a decrease in low frequency and increase in high frequency with SNS, corresponding to increased vagal tone. Additionally, plasma pancreatic peptide was increased and norepinephrine was decreased after SNS in colitis while colon tissue acetylcholine was increased with SNS. This is the first study to the best of our knowledge that demonstrates the benefit of SNS with autonomic mediation. SNS alters the expression of inflammatory cytokines and macrophages as well as modulates neurotransmitters involved in systemic inflammation.
Collapse
Affiliation(s)
| | - Han Zhang
- Division of Gastroenterology and HepatologyJohns Hopkins School of MedicineBaltimoreMDUSA
| | - Nina Zhang
- Division of Gastroenterology and HepatologyJohns Hopkins School of MedicineBaltimoreMDUSA
| | - Jiande D. Z. Chen
- Division of Gastroenterology and HepatologyJohns Hopkins School of MedicineBaltimoreMDUSA
| |
Collapse
|
20
|
Spear ET, Mawe GM. Enteric neuroplasticity and dysmotility in inflammatory disease: key players and possible therapeutic targets. Am J Physiol Gastrointest Liver Physiol 2019; 317:G853-G861. [PMID: 31604034 PMCID: PMC6962496 DOI: 10.1152/ajpgi.00206.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal functions, including motility and secretion, are locally controlled by enteric neural networks housed within the wall of the gut. The fidelity of these functions depends on the precision of intercellular signaling among cellular elements, including enteric neurons, epithelial cells, immune cells, and glia, all of which are vulnerable to disruptive influences during inflammatory events. This review article describes current knowledge regarding inflammation-induced neuroplasticity along key elements of enteric neural circuits, what is known about the causes of these changes, and possible therapeutic targets for protecting and/or repairing the integrity of intrinsic enteric neurotransmission. Changes that have been detected in response to inflammation include increased epithelial serotonin availability, hyperexcitability of intrinsic primary afferent neurons, facilitation of synaptic activity among enteric neurons, and attenuated purinergic neuromuscular transmission. Dysfunctional propulsive motility has been detected in models of colitis, where causes include the changes described above, and in models of multiple sclerosis and other autoimmune conditions, where autoantibodies are thought to mediate dysmotility. Other cells implicated in inflammation-induced neuroplasticity include muscularis macrophages and enteric glia. Targeted treatments that are discussed include 5-hydroxytryptamine receptor 4 agonists, cyclooxygenase inhibitors, antioxidants, B cell depletion therapy, and activation of anti-inflammatory pathways.
Collapse
Affiliation(s)
- Estelle T. Spear
- 1Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, Stanford University, Stanford, California
| | - Gary M. Mawe
- 2Department of Neurological Sciences, The University of Vermont, Burlington, Vermont
| |
Collapse
|
21
|
Aho Fält U, Lindsten M, Strandberg S, Dahlberg M, Butt S, Nilsson E, Zawadzki A, Johnson LB. Percutaneous tibial nerve stimulation (PTNS): an alternative treatment option for chronic therapy resistant anal fissure. Tech Coloproctol 2019; 23:361-365. [PMID: 30972649 PMCID: PMC6536470 DOI: 10.1007/s10151-019-01972-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/26/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of the present study was to evaluate percutaneous tibial nerve stimulation (PTNS) for treatment resistant chronic anal fissure. METHODS Consecutive patients with chronic anal fissure were treated with neuromodulation via the posterior tibial nerve between October 2013 and January 2014. Patients had PTNS for 30 min on 10 consecutive days. All patients had failed conventional medical treatment. The visual analogue scale (VAS) score, St. Marks score, Wexner's constipation score, Brief Pain Inventory (BPI-SF), bleeding and mucosal healing were evaluated before treatment, at termination, after 3 months, and then yearly for 3 years. RESULTS Ten patients (4 males and 6 females; mean age 49.8 years) were identified but only 9 were evaluated as one patient's fissure healed before PTNS was started. At 3-year follow-up, fissures had remained completely healed in 5 out of 9 patients. All patients stopped bleeding and were almost completely pain-free at 3 years (VAS p = 0.010) and pain relief improved from 50% at completion to 90% at 3 years. The patients' Wexner constipation scores improved significantly (p = 0.007). CONCLUSIONS In this small series, PTNS enhanced healing of chronic anal fissure and reduced pain and bleeding with an associated improvement in bowel function.
Collapse
Affiliation(s)
- Ursula Aho Fält
- Department of Surgery, Pelvic Floor Centre, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Martin Lindsten
- Department of Surgery, Pelvic Floor Centre, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Sara Strandberg
- Department of Surgery, Pelvic Floor Centre, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Mari Dahlberg
- Department of Surgery, Pelvic Floor Centre, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Salma Butt
- Department of Surgery, Institution of Clinical Sciences Malmö, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Emelie Nilsson
- Department of Surgery, Institution of Clinical Sciences Malmö, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Antoni Zawadzki
- Department of Surgery, Pelvic Floor Centre, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden
| | - Louis Banka Johnson
- Department of Surgery, Pelvic Floor Centre, Skåne University Hospital-SUS, Lund University, 205 02, Malmö, Sweden.
| |
Collapse
|
22
|
González-González M, Díaz-Zepeda C, Eyzaguirre-Velásquez J, González-Arancibia C, Bravo JA, Julio-Pieper M. Investigating Gut Permeability in Animal Models of Disease. Front Physiol 2019; 9:1962. [PMID: 30697168 PMCID: PMC6341294 DOI: 10.3389/fphys.2018.01962] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/31/2018] [Indexed: 12/12/2022] Open
Abstract
A growing number of investigations report the association between gut permeability and intestinal or extra-intestinal disorders under the basis that translocation of gut luminal contents could affect tissue function, either directly or indirectly. Still, in many cases it is unknown whether disruption of the gut barrier is a causative agent or a consequence of these conditions. Adequate experimental models are therefore required to further understand the pathophysiology of health disorders associated to gut barrier disruption and to develop and test pharmacological treatments. Here, we review the current animal models that display enhanced intestinal permeability, and discuss (1) their suitability to address mechanistic questions, such as the association between gut barrier alterations and disease and (2) their validity to test potential treatments for pathologies that are characterized by enhanced intestinal permeability.
Collapse
Affiliation(s)
- Marianela González-González
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camilo Díaz-Zepeda
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Johana Eyzaguirre-Velásquez
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Camila González-Arancibia
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Javier A Bravo
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Marcela Julio-Pieper
- Grupo de NeuroGastroBioquímica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| |
Collapse
|
23
|
Hvas CL, Bendix M, Dige A, Dahlerup JF, Agnholt J. Current, experimental, and future treatments in inflammatory bowel disease: a clinical review. Immunopharmacol Immunotoxicol 2018; 40:446-460. [PMID: 29745777 DOI: 10.1080/08923973.2018.1469144] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel diseases (IBDs) may result from dysregulated mucosal immune responses directed toward the resident intestinal microbiota. This review describes the hallmark immunobiology of Crohn's disease and ulcerative colitis as well as therapeutic targets and mechanisms of action for current, experimental, and future treatments in IBD. Conventional therapies include 5-aminosalicylic acid, glucocorticosteroids, thiopurines, and methotrexate. Since 1997, monoclonal antibodies have gained widespread use. These consist of antibodies directed against pro-inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)-12, and IL-23, or anti-homing antibodies directed against α4β7 integrin. Emerging oral therapies include modulators of intracellular signal transduction such as Janus kinase inhibitors. Vitamin D may help to regulate innate and adaptive immune responses. Modulation of the intestinal microbiota, using live microorganisms (probiotics), substrates for the colonic microbiota (prebiotics), or fecal microbiota transplantation (FMT), is in development. Dietary supplements are in widespread use, but providing evidence for their benefit is challenging. Stem cell treatment and nervous stimulation are promising future treatments.
Collapse
Affiliation(s)
- Christian L Hvas
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Mia Bendix
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark.,b Medical Department, Randers Regional Hospital , Randers , Denmark
| | - Anders Dige
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Jens F Dahlerup
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| | - Jørgen Agnholt
- a Department of Hepatology and Gastroenterology , Aarhus University Hospital , Aarhus C , Denmark
| |
Collapse
|
24
|
Bonaz B. Is-there a place for vagus nerve stimulation in inflammatory bowel diseases? Bioelectron Med 2018; 4:4. [PMID: 32232080 PMCID: PMC7098256 DOI: 10.1186/s42234-018-0004-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 02/27/2018] [Indexed: 12/11/2022] Open
Abstract
The vagus nerve (VN), the longest nerve of the organism that innervates the gastrointestinal tract, is a mixed nerve composed of 80% of afferent and 20% of efferent fibers. The VN has anti-inflammatory properties, in particular an anti-TNFα effect through the cholinergic anti-inflammatory pathway. The VN is a key component of the autonomic nervous system, i.e. the parasympathetic nervous system. An imbalance of the autonomic nervous system, as represented by a low vagal tone, is described in many diseases and has a pro-inflammatory role. Inflammatory bowel diseases (IBD) are chronic disorders of the gastro-intestinal tract where TNFα is a key cytokine. VN stimulation (VNS), classically used for the treatment of drug resistant epilepsy and depression, would be of interest in the treatment of IBD. We have recently reported in a 6 month follow-up pilot study that VNS improves active Crohn’s disease. Preliminary data of another pilot study confirm this interest. Similarly, VNS has recently been reported to improve rheumatoid arthritis, another TNFα mediated disease. Bioelectronic Medicine, as represented by VNS, opens new therapeutic avenues in the treatment of such chronic inflammatory disorders. In the present manuscript, we will focus on the interest of VNS in IBD.
Collapse
Affiliation(s)
- Bruno Bonaz
- 1Division of Hepato-Gastroenterology, University Hospital, Alpes, F-38000 Grenoble, France.,University Grenoble Alpes, Grenoble Institute of Neurosciences, GIN, Inserm U1216, F-38000 Grenoble, France.,3Division of Hepato-Gastroenterology, CHU Grenoble Alpes, -10217, 38043 Grenoble Cedex 09, CS France
| |
Collapse
|
25
|
Costantini TW, Baird A. Lost your nerve? Modulating the parasympathetic nervous system to treat inflammatory bowel disease. J Physiol 2017; 594:4097-8. [PMID: 27477606 DOI: 10.1113/jp272372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/15/2016] [Indexed: 12/23/2022] Open
Affiliation(s)
- Todd W Costantini
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, UC San Diego Health, San Diego, CA, USA
| | - Andrew Baird
- Division of Trauma, Surgical Critical Care, Burns and Acute Care Surgery, Department of Surgery, UC San Diego Health, San Diego, CA, USA
| |
Collapse
|
26
|
Etchepare N, Bregeon J, Quénéhervé L, Haddara S, Touchefeu Y, Neunlist M, Coron E. Development of a porcine model for assessment of mucosal repair following endoscopic resection of the lower gastrointestinal tract. Endosc Int Open 2017; 5:E1014-E1019. [PMID: 29159277 PMCID: PMC5634855 DOI: 10.1055/s-0043-115383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND AIMS Endoscopic mucosal resection (EMR) is widely performed for the treatment of colorectal polyps. However, the pathophysiological mechanisms of mucosal repair, including in situations at high risk of post-polypectomy bleeding, remain largely unknown. The objective of our study was to develop a porcine model of EMR in the lower gastrointestinal tract to monitor mucosal wound healing over time. METHODS Under general anesthesia, five large wounds were created in the lower gastrointestinal tract at different times, i. e. at day 0, 3, 7, 10, and 14, by multiband EMR, in each of the six pigs in the study. A colorectal resection was performed at day 14 and the animal euthanized. Repeated endoscopic and endomicroscopic examination, and histological analysis were performed. RESULTS No complications occurred and all animals reached the study end point. The endoscopic aspect of wound healing evolved into different phases with first a fibrin deposit covering the wounds which then gave way to granulomatous tissue. The size of the wound regressed significantly as early as day 3. Re-epithelialization of the wound started from day 7, and neo-mucosal crypts appeared from day 10. The endomicroscopic analysis described a 'ground glass appearance' from day 3 and irregular crypts from day 10, which was consistent with histological data. Good agreement between macroscopic, endomicroscopic, and histological parameters of mucosal wound healing was observed in vivo. CONCLUSION This study demonstrates for the first time the feasibility of an experimental in vivo porcine model of lower gastrointestinal endoscopic resections to monitor tissue repair. This model might be helpful to document pharmacological approaches for preventing complications of endoscopic procedures performed in humans.
Collapse
Affiliation(s)
- Nicolas Etchepare
- Institut des Maladies de l’Appareil Digestif, Hôtel Dieu, Centre Hospitalier Universitaire, Nantes, France
| | - Jérémy Bregeon
- INSERM U 1235 – TENS, Université de Nantes, Faculté de Médecine, Nantes, France
| | - Lucille Quénéhervé
- Institut des Maladies de l’Appareil Digestif, Hôtel Dieu, Centre Hospitalier Universitaire, Nantes, France
| | - Sami Haddara
- Institut des Maladies de l’Appareil Digestif, Hôtel Dieu, Centre Hospitalier Universitaire, Nantes, France
| | - Yann Touchefeu
- Institut des Maladies de l’Appareil Digestif, Hôtel Dieu, Centre Hospitalier Universitaire, Nantes, France
| | - Michel Neunlist
- INSERM U 1235 – TENS, Université de Nantes, Faculté de Médecine, Nantes, France
| | - Emmanuel Coron
- Institut des Maladies de l’Appareil Digestif, Hôtel Dieu, Centre Hospitalier Universitaire, Nantes, France,INSERM U 1235 – TENS, Université de Nantes, Faculté de Médecine, Nantes, France,Corresponding author Emmanuel Coron Institut des Maladies de l’Appareil DigestifCHU de Nantes1 place Alexis Ricordeau44093 Nantes Cedex 1France+33-2-40083333
| |
Collapse
|
27
|
Vanhaecke T, Aubert P, Grohard PA, Durand T, Hulin P, Paul-Gilloteaux P, Fournier A, Docagne F, Ligneul A, Fressange-Mazda C, Naveilhan P, Boudin H, Le Ruyet P, Neunlist M. L. fermentum CECT 5716 prevents stress-induced intestinal barrier dysfunction in newborn rats. Neurogastroenterol Motil 2017; 29. [PMID: 28370715 DOI: 10.1111/nmo.13069] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal epithelial barrier (IEB) dysfunction plays a critical role in various intestinal disorders affecting infants and children, including the development of food allergies and colitis. Recent studies highlighted the role of probiotics in regulating IEB functions and behavior in adults, but their effects in the newborn remain largely unknown. We therefore characterized in rat pups, the impact of Lactobacillus fermentum CECT 5716 (L. fermentum) on stress-induced IEB dysfunction, systemic immune response and exploratory behavior. METHODS Newborn rats received daily by gavage either L. fermentum or water. Intestinal permeability to fluorescein sulfonic acid (FSA) and horseradish peroxidase (HRP) was measured following maternal separation (MS) and water avoidance stress (WAS). Immunohistochemical, transcriptomic, and Western blot analysis of zonula occludens-1 (ZO-1) distribution and expression were performed. Anxiety-like and exploratory behavior was assessed using the elevated plus maze test. Cytokine secretion of activated splenocytes was also evaluated. KEY RESULTS L. fermentum prevented MS and WAS-induced IEB dysfunction in vivo. L. fermentum reduced permeability to both FSA and HRP in the small intestine but not in the colon. L. fermentum increased expression of ZO-1 and prevented WAS-induced ZO-1 disorganization in ileal epithelial cells. L. fermentum also significantly reduced stress-induced increase in plasma corticosteronemia. In activated splenocytes, L. fermentum enhanced IFNγ secretion while it prevented IL-4 secretion. Finally, L. fermentum increased exploratory behavior. CONCLUSIONS & INFERENCES These results suggest that L. fermentum could provide a novel tool for the prevention and/or treatment of gastrointestinal disorders associated with altered IEB functions in the newborn.
Collapse
Affiliation(s)
- T Vanhaecke
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France.,Lactalis Recherche et Développement, Retiers, France
| | - P Aubert
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - P-A Grohard
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - T Durand
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - P Hulin
- Université de Nantes, Nantes, France.,MicroPICell - Cellular and Tissular Imaging Core Facility of Nantes, SFR Santé F. Bonamy-FED 4203/Inserm UMS016/CNRS UMS3556, Nantes, France
| | - P Paul-Gilloteaux
- Université de Nantes, Nantes, France.,MicroPICell - Cellular and Tissular Imaging Core Facility of Nantes, SFR Santé F. Bonamy-FED 4203/Inserm UMS016/CNRS UMS3556, Nantes, France
| | - A Fournier
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders (PhIND), Centre Cyceron, Caen, France
| | - F Docagne
- Normandie Univ, UNICAEN, INSERM, Physiopathology and Imaging of Neurological Disorders (PhIND), Centre Cyceron, Caen, France
| | - A Ligneul
- Lactalis Recherche et Développement, Retiers, France
| | | | - P Naveilhan
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - H Boudin
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| | - P Le Ruyet
- Lactalis Recherche et Développement, Retiers, France
| | - M Neunlist
- INSERM U1235, Nantes, France.,Université de Nantes, Nantes, France.,Institut des Maladies de l'Appareil Digestif, Nantes, France
| |
Collapse
|