1
|
Stavely R, Robinson AM, Fraser S, Filippone RT, Stojanovska V, Eri R, Apostolopoulos V, Sakkal S, Nurgali K. Bone marrow-derived mesenchymal stem cells mitigate chronic colitis and enteric neuropathy via anti-inflammatory and anti-oxidative mechanisms. Sci Rep 2024; 14:6649. [PMID: 38503815 PMCID: PMC10951223 DOI: 10.1038/s41598-024-57070-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 03/14/2024] [Indexed: 03/21/2024] Open
Abstract
Current treatments for inflammatory bowel disease (IBD) are often inadequate due to limited efficacy and toxicity, leading to surgical resection in refractory cases. IBD's broad and complex pathogenesis involving the immune system, enteric nervous system, microbiome, and oxidative stress requires more effective therapeutic strategies. In this study, we investigated the therapeutic potential of bone marrow-derived mesenchymal stem cell (BM-MSC) treatments in spontaneous chronic colitis using the Winnie mouse model which closely replicates the presentation and inflammatory profile of ulcerative colitis. The 14-day BM-MSC treatment regimen reduced the severity of colitis, leading to the attenuation of diarrheal symptoms and recovery in body mass. Morphological and histological abnormalities in the colon were also alleviated. Transcriptomic analysis demonstrated that BM-MSC treatment led to alterations in gene expression profiles primarily downregulating genes related to inflammation, including pro-inflammatory cytokines, chemokines and other biomarkers of inflammation. Further evaluation of immune cell populations using immunohistochemistry revealed a reduction in leukocyte infiltration upon BM-MSC treatment. Notably, enteric neuronal gene signatures were the most impacted by BM-MSC treatment, which correlated with the restoration of neuronal density in the myenteric ganglia. Moreover, BM-MSCs exhibited neuroprotective effects against oxidative stress-induced neuronal loss through antioxidant mechanisms, including the reduction of mitochondrial-derived superoxide and attenuation of oxidative stress-induced HMGB1 translocation, potentially relying on MSC-derived SOD1. These findings suggest that BM-MSCs hold promise as a therapeutic intervention to mitigate chronic colitis by exerting anti-inflammatory effects and protecting the enteric nervous system from oxidative stress-induced damage.
Collapse
Affiliation(s)
- Rhian Stavely
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Ainsley M Robinson
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | | | - Vanesa Stojanovska
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Rajaraman Eri
- School of Science, STEM College, RMIT University, Melbourne, VIC, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.
- Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia.
- Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia.
- Enteric Neuropathy Lab, Western Centre for Health, Research and Education, St Albans, VIC, 3021, Australia.
| |
Collapse
|
2
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
3
|
Stavely R, Abalo R, Nurgali K. Targeting Enteric Neurons and Plexitis for the Management of Inflammatory Bowel Disease. Curr Drug Targets 2021; 21:1428-1439. [PMID: 32416686 DOI: 10.2174/1389450121666200516173242] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/11/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
Abstract
Ulcerative colitis (UC) and Crohn's disease (CD) are pathological conditions with an unknown aetiology that are characterised by severe inflammation of the intestinal tract and collectively referred to as inflammatory bowel disease (IBD). Current treatments are mostly ineffective due to their limited efficacy or toxicity, necessitating surgical resection of the affected bowel. The management of IBD is hindered by a lack of prognostic markers for clinical inflammatory relapse. Intestinal inflammation associates with the infiltration of immune cells (leukocytes) into, or surrounding the neuronal ganglia of the enteric nervous system (ENS) termed plexitis or ganglionitis. Histological observation of plexitis in unaffected intestinal regions is emerging as a vital predictive marker for IBD relapses. Plexitis associates with alterations to the structure, cellular composition, molecular expression and electrophysiological function of enteric neurons. Moreover, plexitis often occurs before the onset of gross clinical inflammation, which may indicate that plexitis can contribute to the progression of intestinal inflammation. In this review, the bilateral relationships between the ENS and inflammation are discussed. These include the effects and mechanisms of inflammation-induced enteric neuronal loss and plasticity. Additionally, the role of enteric neurons in preventing antigenic/pathogenic insult and immunomodulation is explored. While all current treatments target the inflammatory pathology of IBD, interventions that protect the ENS may offer an alternative avenue for therapeutic intervention.
Collapse
Affiliation(s)
- Rhian Stavely
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA,Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Raquel Abalo
- Área de Farmacología y Nutrición, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos (URJC), 28922 Alcorcón, Spain,Unidad Asociada I+D+i del Instituto de Química Médica (IQM), Consejo Superior de Investigaciones Científicas
(CSIC), Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System NeuGut-URJC
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences,
The University of Melbourne, Melbourne, Victoria, Australia,Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Li ZS, Hung LY, Margolis KG, Ambron RT, Sung YJ, Gershon MD. The α isoform of cGMP-dependent protein kinase 1 (PKG1α) is expressed and functionally important in intrinsic primary afferent neurons of the guinea pig enteric nervous system. Neurogastroenterol Motil 2021; 33:e14100. [PMID: 33655600 PMCID: PMC8681866 DOI: 10.1111/nmo.14100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intrinsic primary afferent neurons (IPANs) enable the gut to manifest reflexes in the absence of CNS input. PKG1α is selectively expressed in a subset of neurons in dorsal root ganglia (DRG) and has been linked to nociception and long-term hyperexcitability. METHODS We used immunoblotting, immunocytochemistry, and in vitro assays of IPAN-dependent enteric functions to test hypotheses that subsets of primary neurons of the ENS and DRG share a reliance on PKG1α expression. KEY RESULTS PKG1α immunoreactivity was demonstrated in immunoblots from isolated myenteric ganglia. PKG1α, but not PKG1β, immunoreactivity, was coincident with that of neuronal markers (HuC/D; β3-tubulin) in both enteric plexuses. PKG1α immunoreactivity also co-localized with the immunoreactivities of the IPAN markers, calbindin (100%; myenteric plexus) and cytoplasmic NeuN (98 ± 1% submucosal plexus). CGRP-immunoreactive DRG neurons, identified as visceral afferents by retrograde transport, were PKG1α-immunoreactive. We used intraluminal cholera toxin to determine whether PKG1α was necessary to enable stimulation of the mucosa to activate Fos in enteric neurons. Tetrodotoxin (1.0 µM), low Ca2+ /high Mg2+ media, and the PKG inhibitor, N46 (100 µM), all inhibited Fos activation in myenteric neurons. N46 also concentration dependently inhibited peristaltic reflexes in isolated preparations of distal colon (IC50 = 83.3 ± 1.3 µM). CONCLUSIONS & INFERENCES These data suggest that PKG1α is present and functionally important in IPANs and visceral afferent nociceptive neurons.
Collapse
Affiliation(s)
- Zhi S. Li
- Departments of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | - Lin Y. Hung
- Departments of Pediatrics, Columbia University, New York, NY, USA
| | - Kara G. Margolis
- Departments of Pediatrics, Columbia University, New York, NY, USA
| | - Richard T. Ambron
- Departments of Pathology & Cell Biology, Columbia University, New York, NY, USA
| | - Ying J. Sung
- Departments of Basic Science, The Commonwealth Medical College, Scranton, PA, USA
| | - Michael D. Gershon
- Departments of Pathology & Cell Biology, Columbia University, New York, NY, USA
| |
Collapse
|
5
|
Maria-Ferreira D, Dallazen JL, Corso CR, Nascimento AM, Cipriani TR, da Silva Watanabe P, de Mello Gonçales Sant'Ana D, Baggio CH, de Paula Werner MF. Rhamnogalacturonan polysaccharide inhibits inflammation and oxidative stress and alleviates visceral pain. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
6
|
Schemann M, Frieling T, Enck P. To learn, to remember, to forget-How smart is the gut? Acta Physiol (Oxf) 2020; 228:e13296. [PMID: 31063665 DOI: 10.1111/apha.13296] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/29/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022]
Abstract
The enteric nervous system (ENS) resides within the gut wall and autonomously controls gut functions through coordinated activation of sensory, inter and motor neurons. Its activity is modulated by the enteric immune and endocrine system as well as by afferent and efferent nerves of the parasympathetic and sympathetic nervous system. The ENS is often referred to as the second brain and hence is able to perform sophisticated tasks. We review the evidence that the "smartness" of the ENS may even extend to its ability to learn and to memorize. Examples for habituation, sensitization, conditioned behaviour and long-term facilitation are evidence for various forms of implicit learning. Moreover, we discuss how this may change not only basic Neurogastroenterology but also our understanding of development of gut diseases and chronic disorders in gut functions. At the same time, we identify open questions and future challenges to confirm learning, memory and memory deficits in the gut. Despite some remaining experimental challenges, we are convinced that the gut is able to learn and are tempted to answer the question with: Yes, the gut is smart.
Collapse
Affiliation(s)
| | | | - Paul Enck
- Department of Internal Medicine VI, Psychosomatic Medicine and Psychotherapy University Hospital Tübingen Tübingen Germany
| |
Collapse
|
7
|
Filippone RT, Robinson AM, Jovanovska V, Stavely R, Apostolopoulos V, Bornstein JC, Nurgali K. Targeting eotaxin-1 and CCR3 receptor alleviates enteric neuropathy and colonic dysfunction in TNBS-induced colitis in guinea pigs. Neurogastroenterol Motil 2018; 30:e13391. [PMID: 29968270 DOI: 10.1111/nmo.13391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 05/14/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND The accumulation of eosinophils is mediated by the chemokine receptor-3 (CCR3)-eotaxin axis. Increased expression of eotaxin and its receptor is associated with inflammatory bowel disease (IBD). Activation of eosinophils causes the release of cationic proteins that are neurotoxic such as eosinophil-derived neurotoxin (EDN). Damage to enteric neurons alters neurally controlled functions of the gut correlated with intestinal inflammation. We hypothesized that inhibition of the CCR3-eotaxin axis will prevent inflammation-induced functional changes to the gastrointestinal tract. METHODS Hartley guinea pigs were administered with trinitrobenzene sulfonate (TNBS; 30 mg/kg in 30% ethanol) intrarectally to induce colitis. A CCR3 receptor antagonist (SB 328437 [SB3]) was injected intraperitoneally 1 hour postinduction of colitis. Animals were euthanized 7 days post-treatment and colon tissues were collected for ex vivo studies. The EDN-positive eosinophils in the colon, indicating eosinophil activation, were quantified by immunohistochemistry. Effects of SB3 treatment on gross morphological damage, enteric neuropathy, and colonic dysmotility were determined by histology, immunohistochemistry, and organ bath experiments. KEY RESULTS The number of EDN-positive eosinophils was significantly increased in the lamina propria in close proximity to myenteric ganglia in inflamed colon. The TNBS-induced inflammation caused significant damage to colonic architecture and inhibition of colonic motility. Treatment with SB3 antagonist attenuated inflammation-associated morphological damage in the colon, reduced infiltration of EDN-positive eosinophils and restored colonic motility to levels comparable to control and sham-treated guinea pigs. CONCLUSION & INFERENCES This is the first study demonstrating that inhibition of CCR3-eotaxin axis alleviates enteric neuropathy and restores functional changes in the gut associated with TNBS-induced colitis.
Collapse
Affiliation(s)
- R T Filippone
- College of Health and Biomedicine, Victoria University, Melbourne, Vic., Australia.,Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - A M Robinson
- College of Health and Biomedicine, Victoria University, Melbourne, Vic., Australia.,Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - V Jovanovska
- College of Health and Biomedicine, Victoria University, Melbourne, Vic., Australia.,Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - R Stavely
- College of Health and Biomedicine, Victoria University, Melbourne, Vic., Australia.,Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - V Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia
| | - J C Bornstein
- Department of Physiology, Melbourne University, Melbourne, Vic., Australia
| | - K Nurgali
- College of Health and Biomedicine, Victoria University, Melbourne, Vic., Australia.,Institute for Health and Sport, Victoria University, Melbourne, Vic., Australia.,Regenerative, Medicine and Stem Cells Program, Department of Medicine Western Health, Melbourne University, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, Vic., Australia
| |
Collapse
|
8
|
Robinson AM, Rahman AA, Miller S, Stavely R, Sakkal S, Nurgali K. The neuroprotective effects of human bone marrow mesenchymal stem cells are dose-dependent in TNBS colitis. Stem Cell Res Ther 2017; 8:87. [PMID: 28420434 PMCID: PMC5395912 DOI: 10.1186/s13287-017-0540-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/14/2017] [Accepted: 03/17/2017] [Indexed: 02/08/2023] Open
Abstract
Background The incidence of inflammatory bowel diseases (IBD) is increasing worldwide with patients experiencing severe impacts on their quality of life. It is well accepted that intestinal inflammation associates with extensive damage to the enteric nervous system (ENS), which intrinsically innervates the gastrointestinal tract and regulates all gut functions. Hence, treatments targeting the enteric neurons are plausible for alleviating IBD and associated complications. Mesenchymal stem cells (MSCs) are gaining wide recognition as a potential therapy for many diseases due to their immunomodulatory and neuroprotective qualities. However, there is a large discrepancy regarding appropriate cell doses used in both clinical trials and experimental models of disease. We have previously demonstrated that human bone marrow MSCs exhibit neuroprotective and anti-inflammatory effects in a guinea-pig model of 2,4,6-trinitrobenzene-sulfonate (TNBS)-induced colitis; but an investigation into whether this response is dose-dependent has not been conducted. Methods Hartley guinea-pigs were administered TNBS or sham treatment intra-rectally. Animals in the MSC treatment groups received either 1 × 105, 1 × 106 or 3 × 106 MSCs by enema 3 hours after induction of colitis. Colon tissues were collected 72 hours after TNBS administration to assess the effects of MSC treatments on the level of inflammation and damage to the ENS by immunohistochemical and histological analyses. Results MSCs administered at a low dose, 1 × 105 cells, had little or no effect on the level of immune cell infiltrate and damage to the colonic innervation was similar to the TNBS group. Treatment with 1 × 106 MSCs decreased the quantity of immune infiltrate and damage to nerve processes in the colonic wall, prevented myenteric neuronal loss and changes in neuronal subpopulations. Treatment with 3 × 106 MSCs had similar effects to 1 × 106 MSC treatments. Conclusions The neuroprotective effect of MSCs in TNBS colitis is dose-dependent. Increasing doses higher than 1 × 106 MSCs demonstrates no further therapeutic benefit than 1 × 106 MSCs in preventing enteric neuropathy associated with intestinal inflammation. Furthermore, we have established an optimal dose of MSCs for future studies investigating intestinal inflammation, the enteric neurons and stem cell therapy in this model.
Collapse
Affiliation(s)
- Ainsley M Robinson
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Ahmed A Rahman
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Sarah Miller
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Rhian Stavely
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Samy Sakkal
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.
| |
Collapse
|
9
|
Robinson AM, Rahman AA, Carbone SE, Randall-Demllo S, Filippone R, Bornstein JC, Eri R, Nurgali K. Alterations of colonic function in the Winnie mouse model of spontaneous chronic colitis. Am J Physiol Gastrointest Liver Physiol 2017; 312:G85-G102. [PMID: 27881401 DOI: 10.1152/ajpgi.00210.2016] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 01/31/2023]
Abstract
UNLABELLED The Winnie mouse, carrying a missense mutation in Muc2, is a model for chronic intestinal inflammation demonstrating symptoms closely resembling inflammatory bowel disease (IBD). Alterations to the immune environment, morphological structure, and innervation of Winnie mouse colon have been identified; however, analyses of intestinal transit and colonic functions have not been conducted. In this study, we investigated in vivo intestinal transit in radiographic studies and in vitro motility of the isolated colon in organ bath experiments. We compared neuromuscular transmission using conventional intracellular recording between distal colon of Winnie and C57BL/6 mice and smooth muscle contractions using force displacement transducers. Chronic inflammation in Winnie mice was confirmed by detection of lipocalin-2 in fecal samples over 4 wk and gross morphological damage to the colon. Colonic transit was faster in Winnie mice. Motility was altered including decreased frequency and increased speed of colonic migrating motor complexes and increased occurrence of short and fragmented contractions. The mechanisms underlying colon dysfunctions in Winnie mice included inhibition of excitatory and fast inhibitory junction potentials, diminished smooth muscle responses to cholinergic and nitrergic stimulation, and increased number of α-smooth muscle actin-immunoreactive cells. We conclude that diminished excitatory responses occur both prejunctionally and postjunctionally and reduced inhibitory purinergic responses are potentially a prejunctional event, while diminished nitrergic inhibitory responses are probably due to a postjunction mechanism in the Winnie mouse colon. Many of these changes are similar to disturbed motor functions in IBD patients indicating that the Winnie mouse is a model highly representative of human IBD. NEW & NOTEWORTHY This is the first study to provide analyses of intestinal transit and whole colon motility in an animal model of spontaneous chronic colitis. We found that cholinergic and purinergic neuromuscular transmission, as well as the smooth muscle cell responses to cholinergic and nitrergic stimulation, is altered in the chronically inflamed Winnie mouse colon. The changes to intestinal transit and colonic function we identified in the Winnie mouse are similar to those seen in inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Ainsley M Robinson
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Ahmed A Rahman
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Simona E Carbone
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Sarron Randall-Demllo
- University of Tasmania, School of Health Sciences, Launceston, Tasmania, Australia; and
| | - Rhiannon Filippone
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Joel C Bornstein
- Department of Physiology, Melbourne University, Melbourne, Victoria, Australia
| | - Rajaraman Eri
- University of Tasmania, School of Health Sciences, Launceston, Tasmania, Australia; and
| | - Kulmira Nurgali
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia;
| |
Collapse
|
10
|
McQuade RM, Stojanovska V, Donald E, Abalo R, Bornstein JC, Nurgali K. Gastrointestinal dysfunction and enteric neurotoxicity following treatment with anticancer chemotherapeutic agent 5-fluorouracil. Neurogastroenterol Motil 2016; 28:1861-1875. [PMID: 27353132 DOI: 10.1111/nmo.12890] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/29/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND The use of the anticancer chemotherapeutic agent 5-fluorouracil (5-FU) is often limited by nausea, vomiting, constipation, and diarrhea; these side-effects persist long after treatment. The effects of 5-FU on enteric neurons have not been studied and may provide insight into the mechanisms underlying 5-FU-induced gastrointestinal dysfunction. METHODS Balb/c mice received intraperitoneal injections of 5-FU (23 mg/kg) 3 times/week for 14 days. Gastrointestinal transit was analysed in vivo prior to and following 3, 7, and 14 days of 5-FU treatment via serial x-ray imaging. Following 14 days of 5-FU administration, colons were collected for assessment of ex vivo colonic motility, gross morphological structure, and immunohistochemical analysis of myenteric neurons. Fecal lipocalin-2 and CD45+ leukocytes in the colon were analysed as markers of intestinal inflammation. KEY RESULTS Short-term administration of 5-FU (3 days) increased gastrointestinal transit, induced acute intestinal inflammation and reduced the proportion of neuronal nitric oxide synthase-immunoreactive neurons. Long-term treatment (7, 14 days) resulted in delayed gastrointestinal transit, inhibition of colonic migrating motor complexes, increased short and fragmented contractions, myenteric neuronal loss and a reduction in the number of ChAT-immunoreactive neurons after the inflammation was resolved. Gross morphological damage to the colon was observed following both short- and long-term 5-FU treatment. CONCLUSIONS & INFERENCES Our results indicate that 5-FU induces accelerated gastrointestinal transit associated with acute intestinal inflammation at day 3 after the start of treatment, which may have led to persistent changes in the ENS observed after days 7 and 14 of treatment contributing to delayed gastrointestinal transit and colonic dysmotility.
Collapse
Affiliation(s)
- R M McQuade
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Western Centre for Health, Research and Education, Sunshine Hospital, St Albans, VIC, Australia
| | - V Stojanovska
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Western Centre for Health, Research and Education, Sunshine Hospital, St Albans, VIC, Australia
| | - E Donald
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Western Centre for Health, Research and Education, Sunshine Hospital, St Albans, VIC, Australia
| | - R Abalo
- Área de Farmacología y Nutrición y Unidad Asociada al Instituto de Química Médica (IQM) y al Instituto de Investigación en Ciencias de la Alimentación (CIAL) del Consejo Superior de Investigaciones Científicas (CSIC), Grupo de Excelencia Investigadora URJC-Banco de Santander-Grupo Multidisciplinar de Investigación y Tratamiento del Dolor (i+DOL), Universidad Rey Juan Carlos, Alcorcón, Madrid, Spain
| | - J C Bornstein
- Department of Physiology, Melbourne University, Melbourne, VIC, Australia
| | - K Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Western Centre for Health, Research and Education, Sunshine Hospital, St Albans, VIC, Australia
| |
Collapse
|
11
|
Stavely R, Robinson AM, Miller S, Boyd R, Sakkal S, Nurgali K. Allogeneic guinea pig mesenchymal stem cells ameliorate neurological changes in experimental colitis. Stem Cell Res Ther 2015; 6:263. [PMID: 26718461 PMCID: PMC4697327 DOI: 10.1186/s13287-015-0254-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/12/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022] Open
Abstract
Background The use of mesenchymal stem cells (MSCs) to treat inflammatory bowel disease (IBD) is of great interest because of their immunomodulatory properties. Damage to the enteric nervous system (ENS) is implicated in IBD pathophysiology and disease progression. The most commonly used model to study inflammation-induced changes to the ENS is 2,4,6-trinitrobenzene-sulfonate acid (TNBS)-induced colitis in guinea pigs; however, no studies using guinea pig MSCs in colitis have been performed. This study aims to isolate and characterise guinea pig MSCs and then test their therapeutic potential for the treatment of enteric neuropathy associated with intestinal inflammation. Methods MSCs from guinea pig bone marrow and adipose tissue were isolated and characterised in vitro. In in vivo experiments, guinea pigs received either TNBS for the induction of colitis or sham treatment by enema. MSCs were administered at a dose of 1 × 106 cells via enema 3 h after the induction of colitis. Colon tissues were collected 24 and 72 h after TNBS administration to assess the level of inflammation and damage to the ENS. The secretion of transforming growth factor-β1 (TGF-β1) was analysed in MSC conditioned medium by flow cytometry. Results Cells isolated from both sources were adherent to plastic, multipotent and expressed some human MSC surface markers. In vitro characterisation revealed distinct differences in growth kinetics, clonogenicity and cell morphology between MSC types. In an in vivo model of TNBS-induced colitis, guinea pig bone marrow MSCs were comparatively more efficacious than adipose tissue MSCs in attenuating weight loss, colonic tissue damage and leukocyte infiltration into the mucosa and myenteric plexus. MSCs from both sources were equally neuroprotective in the amelioration of enteric neuronal loss and changes to the neurochemical coding of neuronal subpopulations. MSCs from both sources secreted TGF-β1 which exerted neuroprotective effects in vitro. Conclusions This study is the first evaluating the functional capacity of guinea pig bone marrow and adipose tissue-derived MSCs and providing evidence of their neuroprotective value in an animal model of colitis. In vitro characteristics of MSCs cannot be extrapolated to their therapeutic efficacy. TGF-β1 released by both types of MSCs might have contributed to the attenuation of enteric neuropathy associated with colitis.
Collapse
Affiliation(s)
- Rhian Stavely
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Ainsley M Robinson
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Sarah Miller
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Richard Boyd
- Department of Anatomy and Developmental Biology, Monash University, 19 Innovation Walk, Clayton, 3800, Victoria, Australia.
| | - Samy Sakkal
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| |
Collapse
|
12
|
Stavely R, Robinson AM, Miller S, Boyd R, Sakkal S, Nurgali K. Human adult stem cells derived from adipose tissue and bone marrow attenuate enteric neuropathy in the guinea-pig model of acute colitis. Stem Cell Res Ther 2015; 6:244. [PMID: 26652292 PMCID: PMC4674993 DOI: 10.1186/s13287-015-0231-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/22/2015] [Accepted: 11/09/2015] [Indexed: 02/08/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have been identified as a viable treatment for inflammatory bowel disease (IBD). MSCs derived from bone marrow (BM-MSCs) have predominated in experimental models whereas the majority of clinical trials have used MSCs derived from adipose tissue (AT-MSCs), thus there is little consensus on the optimal tissue source. The therapeutic efficacies of these MSCs are yet to be compared in context of the underlying dysfunction of the enteric nervous system innervating the gastrointestinal tract concomitant with IBD. This study aims to characterise the in vitro properties of MSCs and compare their in vivo therapeutic potential for the treatment of enteric neuropathy associated with intestinal inflammation. Methods BM-MSCs and AT-MSCs were validated and characterised in vitro. In in vivo experiments, guinea-pigs received either 2,4,6-trinitrobenzene-sulfonate acid (TNBS) for the induction of colitis or sham treatment by enema. MSCs were administered at a dose of 1x106 cells via enema 3 hours after the induction of colitis. Colon tissues were collected 24 and 72 hours after TNBS administration to assess the level of inflammation and damage to the ENS. MSC migration to the myenteric plexus in vivo was elucidated by immunohistochemistry and in vitro using a modified Boyden chamber assay. Results Cells exhibited multipotency and a typical surface immunophenotype for validation as bona fide MSCs. In vitro characterisation revealed distinct differences in growth kinetics, clonogenicity and cell morphology between MSC types. In vivo, BM-MSCs were comparatively more effective than AT-MSCs in attenuating leukocyte infiltration and neuronal loss in the myenteric plexus. MSCs from both sources equally ameliorated body weight loss, gross morphological damage to the colon, changes in the neurochemical coding of neuronal subpopulations and the reduction in density of extrinsic and intrinsic nerve fibres innervating the colon. MSCs from both sources migrated to the myenteric plexus in in vivo colitis and in an in vitro assay. Conclusions These data from in vitro experiments suggest that AT-MSCs are ideal for cellular expansion. However, BM-MSCs were more therapeutic in the treatment of enteric neuropathy and plexitis. These characteristics should be considered when deciding on the MSC tissue source.
Collapse
Affiliation(s)
- Rhian Stavely
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Ainsley M Robinson
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Sarah Miller
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Richard Boyd
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia.
| | - Samy Sakkal
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia.
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, Australia. .,College of Health and Biomedicine, Victoria University, Western Centre for Health Research & Education, 176 Furlong Road, St Albans, 3021, VIC, Australia.
| |
Collapse
|
13
|
Robinson AM, Miller S, Payne N, Boyd R, Sakkal S, Nurgali K. Neuroprotective Potential of Mesenchymal Stem Cell-Based Therapy in Acute Stages of TNBS-Induced Colitis in Guinea-Pigs. PLoS One 2015; 10:e0139023. [PMID: 26397368 PMCID: PMC4580595 DOI: 10.1371/journal.pone.0139023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/07/2015] [Indexed: 12/13/2022] Open
Abstract
Background & Aims The therapeutic benefits of mesenchymal stem cells (MSCs), such as homing ability, multipotent differentiation capacity and secretion of soluble bioactive factors which exert neuroprotective, anti-inflammatory and immunomodulatory properties, have been attributed to attenuation of autoimmune, inflammatory and neurodegenerative disorders. In this study, we aimed to determine the earliest time point at which locally administered MSC-based therapies avert enteric neuronal loss and damage associated with intestinal inflammation in the guinea-pig model of colitis. Methods At 3 hours after induction of colitis by 2,4,6-trinitrobenzene-sulfonate (TNBS), guinea-pigs received either human bone marrow-derived MSCs, conditioned medium (CM), or unconditioned medium by enema into the colon. Colon tissues were collected 6, 24 and 72 hours after administration of TNBS. Effects on body weight, gross morphological damage, immune cell infiltration and myenteric neurons were evaluated. RT-PCR, flow cytometry and antibody array kit were used to identify neurotrophic and neuroprotective factors released by MSCs. Results MSC and CM treatments prevented body weight loss, reduced infiltration of leukocytes into the colon wall and the myenteric plexus, facilitated repair of damaged tissue and nerve fibers, averted myenteric neuronal loss, as well as changes in neuronal subpopulations. The neuroprotective effects of MSC and CM treatments were observed as early as 24 hours after induction of inflammation even though the inflammatory reaction at the level of the myenteric ganglia had not completely subsided. Substantial number of neurotrophic and neuroprotective factors released by MSCs was identified in their secretome. Conclusion MSC-based therapies applied at the acute stages of TNBS-induced colitis start exerting their neuroprotective effects towards enteric neurons by 24 hours post treatment. The neuroprotective efficacy of MSC-based therapies can be exerted independently to their anti-inflammatory effects.
Collapse
Affiliation(s)
- Ainsley M. Robinson
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Sarah Miller
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Natalie Payne
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Richard Boyd
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia
| | - Samy Sakkal
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Kulmira Nurgali
- Centre for Chronic Diseases, College of Health and Biomedicine, Victoria University, Melbourne, Australia
- * E-mail:
| |
Collapse
|
14
|
Robinson AM, Sakkal S, Park A, Jovanovska V, Payne N, Carbone SE, Miller S, Bornstein JC, Bernard C, Boyd R, Nurgali K. Mesenchymal stem cells and conditioned medium avert enteric neuropathy and colon dysfunction in guinea pig TNBS-induced colitis. Am J Physiol Gastrointest Liver Physiol 2014; 307:G1115-29. [PMID: 25301186 DOI: 10.1152/ajpgi.00174.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Damage to the enteric nervous system (ENS) associated with intestinal inflammation may underlie persistent alterations to gut functions, suggesting that enteric neurons are viable targets for novel therapies. Mesenchymal stem cells (MSCs) offer therapeutic benefits for attenuation of neurodegenerative diseases by homing to areas of inflammation and exhibiting neuroprotective, anti-inflammatory, and immunomodulatory properties. In culture, MSCs release soluble bioactive factors promoting neuronal survival and suppressing inflammation suggesting that MSC-conditioned medium (CM) provides essential factors to repair damaged tissues. We investigated whether MSC and CM treatments administered by enema attenuate 2,4,6-trinitrobenzene-sulfonic acid (TNBS)-induced enteric neuropathy and motility dysfunction in the guinea pig colon. Guinea pigs were randomly assigned to experimental groups and received a single application of TNBS (30 mg/kg) followed by 1 × 10(6) human bone marrow-derived MSCs, 300 μl CM, or 300 μl unconditioned medium 3 h later. After 7 days, the effect of these treatments on enteric neurons was assessed by histological, immunohistochemical, and motility analyses. MSC and CM treatments prevented inflammation-associated weight loss and gross morphological damage in the colon; decreased the quantity of immune infiltrate in the colonic wall (P < 0.01) and at the level of the myenteric ganglia (P < 0.001); prevented loss of myenteric neurons (P < 0.05) and damage to nerve processes, changes in ChAT, and nNOS immunoreactivity (P < 0.05); and alleviated inflammation-induced colonic dysmotility (contraction speed; P < 0.001, contractions/min; P < 0.05). These results provide strong evidence that both MSC and CM treatments can effectively prevent damage to the ENS and alleviate gut dysfunction caused by TNBS-induced colitis.
Collapse
Affiliation(s)
- Ainsley M Robinson
- College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Samy Sakkal
- College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Anthony Park
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia
| | | | - Natalie Payne
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia; and
| | - Simona E Carbone
- College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Sarah Miller
- College of Health and Biomedicine, Victoria University, Melbourne, Australia
| | - Joel C Bornstein
- Department of Physiology, Melbourne University, Melbourne, Australia
| | - Claude Bernard
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia; Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia; and
| | - Richard Boyd
- Department of Anatomy and Neuroscience, Monash University, Melbourne, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Victoria University, Melbourne, Australia;
| |
Collapse
|
15
|
Chambers JD, Bornstein JC, Gwynne RM, Koussoulas K, Thomas EA. A detailed, conductance-based computer model of intrinsic sensory neurons of the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2014; 307:G517-32. [PMID: 25012843 DOI: 10.1152/ajpgi.00228.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intrinsic sensory neurons (ISNs) of the enteric nervous system respond to stimuli such as muscle tension, muscle length, distortion of the mucosa, and the chemical content in the lumen. ISNs form recurrent networks that probably drive many intestinal motor patterns and reflexes. ISNs express a large number of voltage- and calcium-gated ion channels, some of which are modified by inflammation or repeated physiological stimuli, but how interactions between different ionic currents in ISNs produce both normal and pathological behaviors in the intestine remains unclear. We constructed a model of ISNs including voltage-gated sodium and potassium channels, N-type calcium channels, big conductance calcium-dependent potassium (BK) channels, calcium-dependent nonspecific cation channels (NSCa), intermediate conductance calcium-dependent potassium (IK) channels, hyperpolarization-activated cation (Ih) channels, and internal calcium dynamics. The model was based on data from the literature and our electrophysiological studies. The model reproduced responses to short or long depolarizing current pulses and responses to long hyperpolarizing current pulses. Sensitivity analysis showed that Ih, IK, NSCa, and BK have the largest influence on the number of action potentials observed during prolonged depolarizations. The model also predicts that changes to the voltage of activation for Ih have a large influence on excitability, but changes to the time constant of activation for Ih have a minor effect. Our model identifies how interactions between different iconic currents influence the excitability of ISNs and highlights an important role for Ih in enteric neuroplasticity resulting from disease.
Collapse
Affiliation(s)
- Jordan D Chambers
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia; and
| | - Joel C Bornstein
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia; and
| | - Rachel M Gwynne
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia; and
| | - Katerina Koussoulas
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia; and
| | - Evan A Thomas
- Florey Neuroscience Institutes, Parkville, Victoria, Australia
| |
Collapse
|
16
|
Nurgali K, Qu Z, Hunne B, Thacker M, Pontell L, Furness JB. Morphological and functional changes in guinea-pig neurons projecting to the ileal mucosa at early stages after inflammatory damage. J Physiol 2010; 589:325-39. [PMID: 21098001 DOI: 10.1113/jphysiol.2010.197707] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In the present study the relationship between tissue damage and changed electro-physiological properties of Dogiel type II myenteric neurons within the first 24 hours after induction of inflammation with trinitrobenzene sulfonate (TNBS) in the guinea-pig ileum was investigated. Treatment with TNBS causes damage to the mucosa, inflammatory responses in the mucosa and enteric ganglia and changes in myenteric neuron properties. Thus we hypothesise that the physiological changes in the myenteric neurons could be due to damage to their mucosal processes or inflammation in the vicinity of cell bodies or the processes. We found an association between hyperexcitability of myenteric Dogiel type II neurons and damage to the mucosa and its innervation at 3 and 24 h, times when there was also an inflammatory reaction. The lack of hyperexcitability in neurons from control tissues in which axons projecting to the mucosa were severed suggests that inflammation may be an important contributing factor to the neuronal hyperexcitability at the acute stage of inflammation. Despite mucosal repair and re-innervation of the mucosa before 7 days after induction of inflammation, neuronal hyperexcitability persists. Although the mechanisms underlying neuronal hyperexcitability at the acute stage of inflammation might be different from those underlying long-term changes in the absence of active inflammation in the ganglia, the persistent changes in neuronal excitability may contribute to post-inflammatory gut dysfunctions.
Collapse
Affiliation(s)
- Kulmira Nurgali
- School of Biomedical and Health Sciences, Victoria University, Melbourne, Victoria, Australia.
| | | | | | | | | | | |
Collapse
|
17
|
Lakhan SE, Kirchgessner A. Neuroinflammation in inflammatory bowel disease. J Neuroinflammation 2010; 7:37. [PMID: 20615234 PMCID: PMC2909178 DOI: 10.1186/1742-2094-7-37] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 07/08/2010] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease is a chronic intestinal inflammatory condition, the pathology of which is incompletely understood. Gut inflammation causes significant changes in neurally controlled gut functions including cramping, abdominal pain, fecal urgency, and explosive diarrhea. These symptoms are caused, at least in part, by prolonged hyperexcitability of enteric neurons that can occur following the resolution of colitis. Mast, enterochromaffin and other immune cells are increased in the colonic mucosa in inflammatory bowel disease and signal the presence of inflammation to the enteric nervous system. Inflammatory mediators include 5-hydroxytryptamine and cytokines, as well as reactive oxygen species and the production of oxidative stress. This review will discuss the effects of inflammation on enteric neural activity and potential therapeutic strategies that target neuroinflammation in the enteric nervous system.
Collapse
Affiliation(s)
- Shaheen E Lakhan
- Global Neuroscience Initiative Foundation, Los Angeles, CA, USA.
| | | |
Collapse
|