1
|
Costa DVS, Thomasi B, Brito GAC, Gulbransen BD, Warren CA. The role of the enteric nervous system in the pathogenesis of Clostridioides difficile infection. Nat Rev Gastroenterol Hepatol 2025:10.1038/s41575-025-01071-x. [PMID: 40404838 DOI: 10.1038/s41575-025-01071-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/24/2025]
Abstract
Clostridioides difficile is the leading cause of antibiotic-associated diarrhoea worldwide. In the USA, C. difficile infection (CDI) is the eighth leading cause for hospital readmission and seventh for mortality among all gastrointestinal disorders. Gastrointestinal dysmotility and/or diarrhoea occurs after the acute phase of CDI, but persistent gastrointestinal dysfunction post-infection supports contributions of neuroplasticity in the enteric nervous system (ENS), which has a key role in regulating intestinal motility and secretion, in the natural course of CDI. Here, our goal is to provide an up-to-date summary of how the ENS and extrinsic innervation of the gut are affected by CDI and how ENS responses contribute to CDI pathogenesis and outcomes. Enteric neurons and glia are targets of C. difficile toxins in humans and in preclinical model, and changes to the ENS and extrinsic innervation contribute to intestinal inflammation, damage and secretory diarrhoea. These findings suggest possible bidirectional interaction between CDI and the ENS. More studies focusing on understanding how various neurotransmitters and mediators released by the ENS and extrinsic neurons modulate immune responses to CDI could provide insight into novel pharmacological approaches to balance the host response, improve the management of CDI and prevent gastrointestinal dysfunction post-infection.
Collapse
Affiliation(s)
- Deiziane V S Costa
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | - Beatriz Thomasi
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Gerly A C Brito
- Department of Morphology, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Cirle A Warren
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Rao M, Gulbransen BD. Enteric Glia. Cold Spring Harb Perspect Biol 2025; 17:a041368. [PMID: 38951022 PMCID: PMC11960695 DOI: 10.1101/cshperspect.a041368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Enteric glia are a unique type of peripheral neuroglia that accompany neurons in the enteric nervous system (ENS) of the digestive tract. The ENS displays integrative neural circuits that are capable of governing moment-to-moment gut functions independent of input from the central nervous system. Enteric glia are interspersed with neurons throughout these intrinsic gut neural circuits and are thought to fulfill complex roles directed at maintaining homeostasis in the neuronal microenvironment and at neuroeffector junctions in the gut. Changes to glial functions contribute to a wide range of gastrointestinal diseases, but the precise roles of enteric glia in gut physiology and pathophysiology are still under examination. This review summarizes current concepts regarding enteric glial development, diversity, and functions in health and disease.
Collapse
Affiliation(s)
- Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
3
|
Gonzales J, Gulbransen BD. The Physiology of Enteric Glia. Annu Rev Physiol 2025; 87:353-380. [PMID: 39546562 DOI: 10.1146/annurev-physiol-022724-105016] [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] [Indexed: 11/17/2024]
Abstract
Enteric glia are the partners of neurons in the enteric nervous system throughout the gastrointestinal tract. Roles fulfilled by enteric glia are diverse and contribute to maintaining intestinal homeostasis through interactions with neurons, immune cells, and the intestinal epithelium. Glial influences optimize physiological gut processes such as intestinal motility and epithelial barrier integrity through actions that regulate the microenvironment of the enteric nervous system, the activity of enteric neurons, intestinal epithelial functions, and immune response. Changes to glial phenotype in disease switch glial functions and contribute to intestinal inflammation, dysmotility, pain, neuroplasticity, and tumorigenesis. This review summarizes current concepts regarding the physiological roles of enteric glial cells and their potential contributions to gut disease. The discussion is focused on recent evidence that suggests important glial contributions to gastrointestinal health and pathophysiology.
Collapse
Affiliation(s)
- Jacques Gonzales
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA;
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA;
| |
Collapse
|
4
|
Liang C, Wei S, Ji Y, Lin J, Jiao W, Li Z, Yan F, Jing X. The role of enteric nervous system and GDNF in depression: Conversation between the brain and the gut. Neurosci Biobehav Rev 2024; 167:105931. [PMID: 39447778 DOI: 10.1016/j.neubiorev.2024.105931] [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: 06/26/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Depression is a debilitating mental disorder that causes a persistent feeling of sadness and loss of interest. Approximately 280 million individuals worldwide suffer from depression by 2023. Despite the heavy medical and social burden imposed by depression, pathophysiology remains incompletely understood. Emerging evidence indicates various bidirectional interplay enable communication between the gut and brain. These interplays provide a link between intestinal and central nervous system as well as feedback from cortical and sensory centers to enteric activities, which also influences physiology and behavior in depression. This review aims to overview the significant role of the enteric nervous system (ENS) in the pathophysiology of depression and gut-brain axis's contribution to depressive disorders. Additionally, we explore the alterations in enteric glia cells (EGCs) and glial cell line-derived neurotrophic factor (GDNF) in depression and their involvement in neuronal support, intestinal homeostasis maintains and immune response activation. Modulating ENS function, EGCs and GDNF level could serve as novel strategies for future antidepressant therapy.
Collapse
Affiliation(s)
- Chuoyi Liang
- School of Nursing, Jinan University, Guangzhou, China
| | - Sijia Wei
- School of Nursing, Jinan University, Guangzhou, China
| | - Yelin Ji
- School of Nursing, Jinan University, Guangzhou, China
| | - Jiayi Lin
- School of Nursing, Jinan University, Guangzhou, China
| | - Wenli Jiao
- School of Nursing, Jinan University, Guangzhou, China
| | - Zhiying Li
- School of Nursing, Jinan University, Guangzhou, China
| | - Fengxia Yan
- School of Nursing, Jinan University, Guangzhou, China.
| | - Xi Jing
- School of Nursing, Jinan University, Guangzhou, China; Guangdong-Hong Kong-Macau Great Bay Area Geoscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
5
|
Reiner S, Linda S, Ebrahim H, Patrick L, Sven W. The role of reactive enteric glia-macrophage interactions in acute and chronic inflammation. Neurogastroenterol Motil 2024:e14947. [PMID: 39428750 DOI: 10.1111/nmo.14947] [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: 03/04/2024] [Revised: 09/18/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Enteric glia are a heterogeneous population of peripheral glia within the enteric nervous system and play pivotal roles in gut homeostasis, tissue integrity, coordination of motility, and intestinal immune responses. Under physiological conditions, they communicate with enteric neurons to control intestinal motility. In contrast, enteric glia undergo reactive changes in response to inflammatory signals during enteric neuroinflammation and participate in immune control. In this state, these glia are called reactive enteric glia, which promote cytokine and chemokine secretion and perpetuate immune cell recruitment, thereby affecting disease progression. Interestingly, reactive glia exhibit a huge plasticity and adapt to or shape the immune environment towards a resolving phenotype during inflammation and neuropathies. Recent studies revealed a bidirectional communication between enteric glia and resident and infiltrating immune cells under healthy conditions and in the context of inflammation-based intestinal disorders and neuropathies. While recent reviews give a superb general overview of enteric glial reactivity, we herein discuss the latest evidence on enteric glial reactivity in two prominent inflammatory conditions: acute postoperative inflammation, resulting in postoperative ileus, and chronic inflammation in inflammatory bowel diseases. We define their plasticity during inflammation and the interplay between reactive enteric glia and intestinal macrophages. Finally, we sketch important questions that should be addressed to clarify further the impact of enteric glial reactivity on intestinal inflammation.
Collapse
Affiliation(s)
| | - Schneider Linda
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Hamza Ebrahim
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Leven Patrick
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| | - Wehner Sven
- Department of Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
6
|
Jamka JR, Gulbransen BD. Mechanisms of enteric neuropathy in diverse contexts of gastrointestinal dysfunction. Neurogastroenterol Motil 2024:e14870. [PMID: 39038157 DOI: 10.1111/nmo.14870] [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: 01/30/2024] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
The enteric nervous system (ENS) commands moment-to-moment gut functions through integrative neurocircuitry housed in the gut wall. The functional continuity of ENS networks is disrupted in enteric neuropathies and contributes to major disturbances in normal gut activities including abnormal gut motility, secretions, pain, immune dysregulation, and disrupted signaling along the gut-brain axis. The conditions under which enteric neuropathy occurs are diverse and the mechanistic underpinnings are incompletely understood. The purpose of this brief review is to summarize the current understanding of the cell types involved, the conditions in which neuropathy occurs, and the mechanisms implicated in enteric neuropathy such as oxidative stress, toll like receptor signaling, purines, and pre-programmed cell death.
Collapse
Affiliation(s)
- Julia R Jamka
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
7
|
Lima FGDM, Silva MPAD, Sestak SS, Guarnier FA, de Oliveira AP, Kuller JV, Gulbransen BD, Perles JVCM, Zanoni JN. Cancer-induced morphological changes in enteric glial cells in the jejunum of Walker-256 tumor-bearing rats. Acta Histochem 2024; 126:152146. [PMID: 38422841 PMCID: PMC11039380 DOI: 10.1016/j.acthis.2024.152146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Cancer-induced cachexia is associated with systemic inflammation and gastrointestinal dysfunction. How changes to cells of the enteric nervous system contribute to gut dysfunction in tumor development and cancer cachexia is unknown. Here, we tested the hypothesis that changes to enteric glia, a type of peripheral glia that surround enteric neurons and regulate gut homeostasis, are associated with tumor development and that supplementing with the antioxidant L-glutathione is protective against the changes induced. Immunohistochemistry for neurons, enteric glial cells and immune cells was performed in whole-mount preparations and frozen histological sections of the jejunum from 20 Wistar rats, distributed in 4 groups: control, tumor of Walker-256, control administered with 1 % L-glutathione, and tumor of Walker-256 administered with 1 % L-glutathione. Morphoquantitative analyses were made using Image-Pro® Plus 4.5 and ImageJ® 1.43° software. Tumor development significantly reduced neuronal and glial cell populations in the myenteric and submucosal plexuses and enlarged glial cell body area in the submucosal plexus. In contrast, tumors increased glia in the jejunal mucosa and this effect was accompanied by B-lymphocyte recruitment. GSH-supplemented diet was not sufficient to protect against changes to neurons and glia in the submucosal plexus but was partially protective in the myenteric plexus. L-glutathione had no effect on physiological parameters of cachexia but was sufficient to preserve enteric glial cell density in the myenteric plexus. These results suggest that changes to both enteric neurons and glia likely contribute to the gastrointestinal effects of tumor development and that oxidative stress contributes to these effects in the enteric nervous system.
Collapse
Affiliation(s)
| | | | - Sabrina Silva Sestak
- Laboratory of Enteric Neural Plasticity, State University of Maringá, Maringá, PR, Brazil
| | | | | | - João Victor Kuller
- Laboratory of Enteric Neural Plasticity, State University of Maringá, Maringá, PR, Brazil
| | | | | | | |
Collapse
|
8
|
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
|
9
|
Czyżewski W, Mazurek M, Sakwa L, Szymoniuk M, Pham J, Pasierb B, Litak J, Czyżewska E, Turek M, Piotrowski B, Torres K, Rola R. Astroglial Cells: Emerging Therapeutic Targets in the Management of Traumatic Brain Injury. Cells 2024; 13:148. [PMID: 38247839 PMCID: PMC10813911 DOI: 10.3390/cells13020148] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic Brain Injury (TBI) represents a significant health concern, necessitating advanced therapeutic interventions. This detailed review explores the critical roles of astrocytes, key cellular constituents of the central nervous system (CNS), in both the pathophysiology and possible rehabilitation of TBI. Following injury, astrocytes exhibit reactive transformations, differentiating into pro-inflammatory (A1) and neuroprotective (A2) phenotypes. This paper elucidates the interactions of astrocytes with neurons, their role in neuroinflammation, and the potential for their therapeutic exploitation. Emphasized strategies encompass the utilization of endocannabinoid and calcium signaling pathways, hormone-based treatments like 17β-estradiol, biological therapies employing anti-HBGB1 monoclonal antibodies, gene therapy targeting Connexin 43, and the innovative technique of astrocyte transplantation as a means to repair damaged neural tissues.
Collapse
Affiliation(s)
- Wojciech Czyżewski
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Marek Mazurek
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| | - Leon Sakwa
- Student Scientific Society, Kazimierz Pulaski University of Radom, 26-600 Radom, Poland;
| | - Michał Szymoniuk
- Student Scientific Association, Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Jennifer Pham
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Barbara Pasierb
- Department of Dermatology, Radom Specialist Hospital, 26-600 Radom, Poland;
| | - Jakub Litak
- Department of Clinical Immunology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Ewa Czyżewska
- Department of Otolaryngology, Mazovian Specialist Hospital, 26-617 Radom, Poland;
| | - Michał Turek
- Student Scientific Society, Medical University of Lublin, 20-954 Lublin, Poland; (J.P.); (M.T.)
| | - Bartłomiej Piotrowski
- Institute of Automatic Control and Robotics, Warsaw University of Technology, 00-661 Warsaw, Poland;
| | - Kamil Torres
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Radosław Rola
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, 20-954 Lublin, Poland; (M.M.); (R.R.)
| |
Collapse
|
10
|
de Oliveira AP, Perles JVCM, de Souza SRG, Sestak SS, da Motta Lima FG, Almeida GHDR, Cicero LR, Clebis NK, Guarnier FA, Blegniski FP, Vasconcelos RC, Araújo AA, Comar JF, Moreira LS, Sehaber-Sierakowski CC, Zanoni KPS, Zanoni JN. L-glutathione 1% promotes neuroprotection of nitrergic neurons and reduces the oxidative stress in the jejunum of rats with Walker-256-bearing tumor. Neurogastroenterol Motil 2023; 35:e14688. [PMID: 37831748 DOI: 10.1111/nmo.14688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/21/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
AIMS Our main goals were to investigate the effects of L-glutathione (1%) treatment in Walker-256 tumor-bearing rats by analyzing immunoreactive neurons (IR), responsive to the nNOS enzyme and 3-Nitrotyrosine, in their jejunum myenteric plexus. Moreover, the oxidative state and inflammatory process in these animals were investigated. METHODS Four experimental groups were utilized: control (C), control treated with L-glutathione (CGT), Walker-256 tumor-bearing rats (TW), and Walker-256 tumor-bearing rats treated with L-glutathione (TWGT). After 14 days of tumor inoculation, the jejunum was collected for immunohistochemical techniques and assessment of oxidative status. Plasma was collected to evaluate oxidative status and measure cytokines. RESULTS The TW group exhibited a decrease of reduced glutathione in their jejunum, which was prevented in the L-glutathione treated TWGT group. TW animals presented pronounced oxidative stress by increasing levels of lipoperoxidation in their jejunum and malondialdehyde in their plasma; however, the L-glutathione treatment in TWGT group was not able to avoid it. The total antioxidant capacity was altered in groups TW and TWGT, yet the last one had a better index in their plasma. The IL-10, and TNF-α levels increased in TWGT animals. The nNOS-IR neuron density decreased in the jejunum myenteric plexus of the TW group, which was avoided in the TWGT group. The nNOS +3-Nitrotyrosine neurons quantification did not show significative alterations. CONCLUSION The treatment with L-glutathione (1%) imposed an important defense to some parameters of oxidative stress induced by TW-256, leading to neuroprotection to the loss in the nNOS-IR neuron density.
Collapse
Affiliation(s)
- Ana Paula de Oliveira
- Department of Physiology Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | - Sabrina Silva Sestak
- Department of Physiology Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | - Lídia Rodrigues Cicero
- Department of Pharmaceutical Sciences, State University of Maringá, Maringá, Paraná, Brazil
| | - Naianne Kelly Clebis
- Department of Morphology, Center of Biosciences Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | | | - Roseane Carvalho Vasconcelos
- Department of Dentistry, Program of Oral pathology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | - Aurigena Antunes Araújo
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Rio Grande do Norte, Brazil
| | | | | | | | | | - Jacqueline Nelisis Zanoni
- Department of Physiology Sciences, State University of Maringá, Maringá, Paraná, Brazil
- Department of Morphology Sciences, State University of Maringá, Maringá, Paraná, Brazil
- Department of Pharmaceutical Sciences, State University of Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
11
|
Stavely R, Ott LC, Rashidi N, Sakkal S, Nurgali K. The Oxidative Stress and Nervous Distress Connection in Gastrointestinal Disorders. Biomolecules 2023; 13:1586. [PMID: 38002268 PMCID: PMC10669114 DOI: 10.3390/biom13111586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress is increasingly recognized as a central player in a range of gastrointestinal (GI) disorders, as well as complications stemming from therapeutic interventions. This article presents an overview of the mechanisms of oxidative stress in GI conditions and highlights a link between oxidative insult and disruption to the enteric nervous system (ENS), which controls GI functions. The dysfunction of the ENS is characteristic of a spectrum of disorders, including neurointestinal diseases and conditions such as inflammatory bowel disease (IBD), diabetic gastroparesis, and chemotherapy-induced GI side effects. Neurons in the ENS, while essential for normal gut function, appear particularly vulnerable to oxidative damage. Mechanistically, oxidative stress in enteric neurons can result from intrinsic nitrosative injury, mitochondrial dysfunction, or inflammation-related pathways. Although antioxidant-based therapies have shown limited efficacy, recognizing the multifaceted role of oxidative stress in GI diseases offers a promising avenue for future interventions. This comprehensive review summarizes the literature to date implicating oxidative stress as a critical player in the pathophysiology of GI disorders, with a focus on its role in ENS injury and dysfunction, and highlights opportunities for the development of targeted therapeutics for these diseases.
Collapse
Affiliation(s)
- Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leah C. Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Niloufar Rashidi
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
- Department of Medicine Western Health, The University of Melbourne, St Albans, VIC 3021, Australia
- Regenerative Medicine and Stem Cell Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, VIC 3021, Australia
| |
Collapse
|
12
|
Sahakian L, Robinson AM, Sahakian L, Stavely R, Kelley MR, Nurgali K. APE1/Ref-1 as a Therapeutic Target for Inflammatory Bowel Disease. Biomolecules 2023; 13:1569. [PMID: 38002251 PMCID: PMC10669584 DOI: 10.3390/biom13111569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic relapsing inflammation of the gastrointestinal tract. The prevalence of IBD is increasing with approximately 4.9 million cases reported worldwide. Current therapies are limited due to the severity of side effects and long-term toxicity, therefore, the development of novel IBD treatments is necessitated. Recent findings support apurinic/apyrimidinic endonuclease 1/reduction-oxidation factor 1 (APE1/Ref-1) as a target in many pathological conditions, including inflammatory diseases, where APE1/Ref-1 regulation of crucial transcription factors impacts significant pathways. Thus, a potential target for a novel IBD therapy is the redox activity of the multifunctional protein APE1/Ref-1. This review elaborates on the status of conventional IBD treatments, the role of an APE1/Ref-1 in intestinal inflammation, and the potential of a small molecule inhibitor of APE1/Ref-1 redox activity to modulate inflammation, oxidative stress response, and enteric neuronal damage in IBD.
Collapse
Affiliation(s)
- Lauren Sahakian
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (L.S.); (A.M.R.)
| | - Ainsley M. Robinson
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (L.S.); (A.M.R.)
| | - Linda Sahakian
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia; (L.S.); (R.S.)
| | - Rhian Stavely
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia; (L.S.); (R.S.)
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Mark R. Kelley
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kulmira Nurgali
- Institute for Health & Sport, Victoria University, Melbourne, VIC 3021, Australia; (L.S.); (A.M.R.)
- Department of Medicine Western Health, The University of Melbourne, Melbourne, VIC 3010, Australia; (L.S.); (R.S.)
- Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| |
Collapse
|
13
|
Scantlen MD, Majd H, Fattahi F. Modeling enteric glia development, physiology and disease using human pluripotent stem cells. Neurosci Lett 2023; 811:137334. [PMID: 37315730 DOI: 10.1016/j.neulet.2023.137334] [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: 02/18/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
Enteric glia play an integral role in many functions of the gastrointestinal (GI) system, but they have not been characterized comprehensively compared to other cells of the gut. Enteric glia are a specialized type of neuroglia in the enteric nervous system (ENS) that support neurons and interact with other cells of the gut such as immune and epithelial cells. The ENS is diffusely spread throughout the GI tract, making it extremely difficult to access and manipulate. As a result, it has remained extremely understudied. Nevertheless, much more is known about enteric neurons than enteric glia despite the glia being 6 times more abundant in humans [1]. In the past two decades, our understanding of enteric glia has greatly expanded and their many roles in the gut have been described and reviewed elsewhere [2-5]. While the field has made substantial progress, there are still a multitude of open questions about enteric glia biology and their role in disease. Many of these questions have remained intractable due to technical limitations of currently available experimental models of the ENS. In this review, we describe the benefits and limitations of the models commonly used to study enteric glia and discuss the ways in which a human pluripotent stem cell (hPSC) derived enteric glia model could help advance the field.
Collapse
Affiliation(s)
- Megan D Scantlen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Homa Majd
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Program in Biomedical Sciences, University of California, San Francisco, San Francisco, CA 94110, USA; Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94110, USA.
| |
Collapse
|
14
|
Dharshika C, Gonzales J, Chow A, Morales-Soto W, Gulbransen BD. Stimulator of interferon genes (STING) expression in the enteric nervous system and contributions of glial STING in disease. Neurogastroenterol Motil 2023; 35:e14553. [PMID: 37309618 PMCID: PMC10266835 DOI: 10.1111/nmo.14553] [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: 10/25/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Appropriate host-microbe interactions are essential for enteric glial development and subsequent gastrointestinal function, but the potential mechanisms of microbe-glial communication are unclear. Here, we tested the hypothesis that enteric glia express the pattern recognition receptor stimulator of interferon genes (STING) and communicate with the microbiome through this pathway to modulate gastrointestinal inflammation. METHODS In situ transcriptional labeling and immunohistochemistry were used to examine STING and IFNβ expression in enteric neurons and glia. Glial-STING KO mice (Sox10CreERT2+/- ;STINGfl/fl ) and IFNβ ELISA were used to characterize the role of enteric glia in canonical STING activation. The role of glial STING in gastrointestinal inflammation was assessed in the 3% DSS colitis model. RESULTS Enteric glia and neurons express STING, but only enteric neurons express IFNβ. While both the myenteric and submucosal plexuses produce IFNβ with STING activation, enteric glial STING plays a minor role in its production and seems more involved in autophagy processes. Furthermore, deleting enteric glial STING does not affect weight loss, colitis severity, or neuronal cell proportions in the DSS colitis model. CONCLUSION Taken together, our data support canonical roles for STING and IFNβ signaling in the enteric nervous system through enteric neurons but that enteric glia do not use these same mechanisms. We propose that enteric glial STING may utilize alternative signaling mechanisms and/or is only active in particular disease conditions. Regardless, this study provides the first glimpse of STING signaling in the enteric nervous system and highlights a potential avenue of neuroglial-microbial communication.
Collapse
Affiliation(s)
- Christine Dharshika
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
- College of Human Medicine, Michigan State University, 804 Service Road, East Lansing, MI, 48824 USA
| | - Jacques Gonzales
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Aaron Chow
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Wilmarie Morales-Soto
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Brian D. Gulbransen
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| |
Collapse
|
15
|
A Comprehensive Analysis of Microflora and Metabolites in the Development of Ulcerative Colitis into Colorectal Cancer Based on the Lung–Gut Correlation Theory. Molecules 2022; 27:molecules27185838. [PMID: 36144573 PMCID: PMC9503129 DOI: 10.3390/molecules27185838] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 12/04/2022] Open
Abstract
The lungs and large intestine can co-regulate inflammation and immunity through the lung–gut axis, in which the transportation of the gut microbiota and metabolites is the most important communication channel. In our previous study, not only did the composition of the gut microbiota and metabolites related to inflammation change significantly during the transition from ulcerative colitis (UC) to colorectal cancer (CRC), but the lung tissues also showed corresponding inflammatory changes, which indicated that gastrointestinal diseases can lead to pulmonary diseases. In order to elucidate the mechanisms of this lung–gut axis, metabolites in bronchoalveolar lavage fluid (BALF) and lung tissues were detected using UHPLC–Q-TOF-MS/MS technology, while microbiome characterization was performed in BALF using 16S rDNA sequencing. The levels of pulmonary metabolites changed greatly during the development of UC to CRC. Among these changes, the concentrations of linoleic acid and 7-hydroxy-3-oxocholic acid gradually increased during the development of UC to CRC. In addition, the composition of the pulmonary microbiota also changed significantly, with an increase in the Proteobacteria and an obvious decrease in the Firmicutes. These changes were consistent with our previous studies of the gut. Collectively, the microbiota and metabolites identified above might be the key markers related to lung and gut diseases, which can be used as an indication of the transition of diseases from the gut to the lung and provide a scientific basis for clinical treatment.
Collapse
|
16
|
Sampath C, Raju AV, Freeman ML, Srinivasan S, Gangula PR. Nrf2 attenuates hyperglycemia-induced nNOS impairment in adult mouse primary enteric neuronal crest cells and normalizes stomach function. Am J Physiol Gastrointest Liver Physiol 2022; 322:G368-G382. [PMID: 35084215 PMCID: PMC8897013 DOI: 10.1152/ajpgi.00323.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Enteric neuronal cells play a vital role in gut motility in humans and experimental rodent models. Patients with diabetes are more vulnerable to gastrointestinal dysfunction due to enteric neuronal degeneration. In this study, we examined the mechanistic role and regulation of nuclear factor-erythroid 2-related factor 2 (Nrf2) in hyperglycemia-induced enteric neuronal cell apoptosis in vitro by using adult mouse primary enteric neuronal crest cells (pENCs). Our data show that hyperglycemia (HG) or inhibition of Nrf2 induces apoptosis by elevating proinflammatory cytokines, reactive oxygen species (ROS) and suppresses neuronal nitric oxide synthase (nNOS-α) via PI3K/Nrf2-mediated signaling. Conversely, treating pENCs with cinnamaldehyde (CNM), a naturally occurring Nrf2 activator, prevented HG-induced apoptosis. These novel data reveal a negative feedback mechanism for GSK-3 activation. To further demonstrate that loss of Nrf2 leads to inflammation, oxidative stress, and reduces nNOS-mediated gastric function, we have used streptozotocin (STZ)-induced diabetic and Nrf2 null female mice. In vivo activation of Nrf2 with CNM (50 mg/kg, 3 days a week, ip) attenuated impaired nitrergic relaxation and delayed gastric emptying (GE) in conventional type 1 diabetic but not in Nrf2 null female mice. Supplementation of CNM normalized diabetes-induced altered gastric antrum protein expression of 1) p-AKT/p-p38MAPK/p-GSK-3β, 2) BH4 (cofactor of nNOS) biosynthesis enzyme GCH-1, 3) nNOSα, 4) TLR4, NF-κB, and 5) inflammatory cytokines (TNF-α, IL-1β, IL-6). We conclude that activation of Nrf2 prevents hyperglycemia-induced apoptosis in pENCs and restores nitrergic-mediated gastric motility and GE in STZ-induced diabetes female mice.NEW & NOTEWORTHY Primary neuronal cell crust (pENCs) in the intestine habitats nNOS and Nrf2, which was suppressed in diabetic gastroparesis. Activation of Nrf2 restored nNOS by suppressing inflammatory markers in pENCs cells. Inhibition of Nrf2 reveals a negative feedback mechanism for the activation of GSK-3. Activation of Nrf2 alleviates STZ-induced delayed gastric emptying and nitrergic relaxation in female mice. Activation of Nrf2 restored impaired gastric BH4 biosynthesis enzyme GCH-1, nNOSα expression thus regulating nitric oxide levels.
Collapse
Affiliation(s)
- Chethan Sampath
- 1Department of ODS and Research, School of Dentistry, Meharry Medical College, Nashville, Tennessee
| | - Abhinav V. Raju
- 2Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia
| | - Michael L. Freeman
- 4Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shanthi Srinivasan
- 2Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia,3Atlanta Veterans Affairs Health Care System, Atlanta, Georgia
| | - Pandu R. Gangula
- 1Department of ODS and Research, School of Dentistry, Meharry Medical College, Nashville, Tennessee
| |
Collapse
|
17
|
Seguella L, Gulbransen BD. Enteric glial biology, intercellular signalling and roles in gastrointestinal disease. Nat Rev Gastroenterol Hepatol 2021; 18:571-587. [PMID: 33731961 PMCID: PMC8324524 DOI: 10.1038/s41575-021-00423-7] [Citation(s) in RCA: 179] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
One of the most transformative developments in neurogastroenterology is the realization that many functions normally attributed to enteric neurons involve interactions with enteric glial cells: a large population of peripheral neuroglia associated with enteric neurons throughout the gastrointestinal tract. The notion that glial cells function solely as passive support cells has been refuted by compelling evidence that demonstrates that enteric glia are important homeostatic cells of the intestine. Active signalling mechanisms between enteric glia and neurons modulate gastrointestinal reflexes and, in certain circumstances, function to drive neuroinflammatory processes that lead to long-term dysfunction. Bidirectional communication between enteric glia and immune cells contributes to gastrointestinal immune homeostasis, and crosstalk between enteric glia and cancer stem cells regulates tumorigenesis. These neuromodulatory and immunomodulatory roles place enteric glia in a unique position to regulate diverse gastrointestinal disease processes. In this Review, we discuss current concepts regarding enteric glial development, heterogeneity and functional roles in gastrointestinal pathophysiology and pathophysiology, with a focus on interactions with neurons and immune cells. We also present a working model to differentiate glial states based on normal function and disease-induced dysfunctions.
Collapse
Affiliation(s)
- Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Brian D Gulbransen
- Department of Physiology, Neuroscience Program, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
18
|
Bjørklund G, Doşa MD, Maes M, Dadar M, Frye RE, Peana M, Chirumbolo S. The impact of glutathione metabolism in autism spectrum disorder. Pharmacol Res 2021; 166:105437. [PMID: 33493659 DOI: 10.1016/j.phrs.2021.105437] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 12/31/2020] [Accepted: 01/12/2021] [Indexed: 12/14/2022]
Abstract
This paper reviews the potential role of glutathione (GSH) in autism spectrum disorder (ASD). GSH plays a key role in the detoxification of xenobiotics and maintenance of balance in intracellular redox pathways. Recent data showed that imbalances in the GSH redox system are an important factor in the pathophysiology of ASD. Furthermore, ASD is accompanied by decreased concentrations of reduced GSH in part caused by oxidation of GSH into glutathione disulfide (GSSG). GSSG can react with protein sulfhydryl (SH) groups, thereby causing proteotoxic stress and other abnormalities in SH-containing enzymes in the brain and blood. Moreover, alterations in the GSH metabolism via its effects on redox-independent mechanisms are other processes associated with the pathophysiology of ASD. GSH-related regulation of glutamate receptors such as the N-methyl-D-aspartate receptor can contribute to glutamate excitotoxicity. Synergistic and antagonistic interactions between glutamate and GSH can result in neuronal dysfunction. These interactions can involve transcription factors of the immune pathway, such as activator protein 1 and nuclear factor (NF)-κB, thereby interacting with neuroinflammatory mechanisms, ultimately leading to neuronal damage. Neuronal apoptosis and mitochondrial dysfunction are recently outlined as significant factors linking GSH impairments with the pathophysiology of ASD. Moreover, GSH regulates the methylation of DNA and modulates epigenetics. Existing data support a protective role of the GSH system in ASD development. Future research should focus on the effects of GSH redox signaling in ASD and should explore new therapeutic approaches by targeting the GSH system.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Toften 24, 8610, Mo i Rana, Norway.
| | - Monica Daniela Doşa
- Department of Pharmacology, Faculty of Medicine, Ovidius University of Constanta, Campus, 900470, Constanta, Romania.
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Richard E Frye
- Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine, Phoenix, AZ, USA
| | | | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy; CONEM Scientific Secretary, Verona, Italy
| |
Collapse
|
19
|
Nyavor Y, Brands CR, Nicholson J, Kuther S, Cox KK, May G, Miller C, Yasuda A, Potter F, Cady J, Heyman HM, Metz TO, Stark TD, Hofmann T, Balemba OB. Supernatants of intestinal luminal contents from mice fed high-fat diet impair intestinal motility by injuring enteric neurons and smooth muscle cells. Neurogastroenterol Motil 2021; 33:e13990. [PMID: 32969549 DOI: 10.1111/nmo.13990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 08/12/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Damage to enteric neurons and impaired gastrointestinal muscle contractions cause motility disorders in 70% of diabetic patients. It is thought that enteric neuropathy and dysmotility occur before overt diabetes, but triggers of these abnormalities are not fully known. We tested the hypothesis that intestinal contents of mice with and without high-fat diet- (HFD-) induced diabetic conditions contain molecules that impair gastrointestinal movements by damaging neurons and disrupting muscle contractions. METHODS Small and large intestinal segments were collected from healthy, standard chow diet (SCD) fed mice. Filtrates of ileocecal contents (ileocecal supernatants; ICS) from HFD or SCD mice were perfused through them. Cultured intact intestinal muscularis externa preparations were used to determine whether ICS and their fractions obtained by solid-phase extraction (SPE) and SPE subfractions collected by high-performance liquid chromatography (HPLC) disrupt muscle contractions by injuring neurons and smooth muscle cells. KEY RESULTS ICS from HFD mice reduced intestinal motility, but those from SCD mice had no effect. ICS, aqueous SPE fractions and two out of twenty HPLC subfractions of aqueous SPE fractions from HFD mice blocked muscle contractions, caused a loss of nitrergic myenteric neurons through inflammation, and reduced smooth muscle excitability. Lipopolysaccharide and palmitate caused a loss of nitrergic myenteric neurons but did not affect muscle contractions. CONCLUSIONS & INFERENCES Unknown molecules in intestinal contents of HFD mice trigger enteric neuropathy and dysmotility. Further studies are required to identify the toxic molecules and their mechanisms of action.
Collapse
Affiliation(s)
- Yvonne Nyavor
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | | | - Jessica Nicholson
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Sydney Kuther
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Kortni K Cox
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - George May
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | | | - Allysha Yasuda
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Forrest Potter
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Joshua Cady
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Heino M Heyman
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Thomas O Metz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Timo D Stark
- Lehrstuhl für Lebensmittelchemie und Molekulare Sensorik, Technische Universität München, Freising, Germany
| | - Thomas Hofmann
- Lehrstuhl für Lebensmittelchemie und Molekulare Sensorik, Technische Universität München, Freising, Germany
| | - Onesmo B Balemba
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| |
Collapse
|
20
|
Sahakian L, Filippone RT, Stavely R, Robinson AM, Yan XS, Abalo R, Eri R, Bornstein JC, Kelley MR, Nurgali K. Inhibition of APE1/Ref-1 Redox Signaling Alleviates Intestinal Dysfunction and Damage to Myenteric Neurons in a Mouse Model of Spontaneous Chronic Colitis. Inflamm Bowel Dis 2020; 27:388-406. [PMID: 32618996 PMCID: PMC8287929 DOI: 10.1093/ibd/izaa161] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) associates with damage to the enteric nervous system (ENS), leading to gastrointestinal (GI) dysfunction. Oxidative stress is important for the pathophysiology of inflammation-induced enteric neuropathy and GI dysfunction. Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a dual functioning protein that is an essential regulator of the cellular response to oxidative stress. In this study, we aimed to determine whether an APE1/Ref-1 redox domain inhibitor, APX3330, alleviates inflammation-induced oxidative stress that leads to enteric neuropathy in the Winnie murine model of spontaneous chronic colitis. METHODS Winnie mice received APX3330 or vehicle via intraperitoneal injections over 2 weeks and were compared with C57BL/6 controls. In vivo disease activity and GI transit were evaluated. Ex vivo experiments were performed to assess functional parameters of colonic motility, immune cell infiltration, and changes to the ENS. RESULTS Targeting APE1/Ref-1 redox activity with APX3330 improved disease severity, reduced immune cell infiltration, restored GI function ,and provided neuroprotective effects to the enteric nervous system. Inhibition of APE1/Ref-1 redox signaling leading to reduced mitochondrial superoxide production, oxidative DNA damage, and translocation of high mobility group box 1 protein (HMGB1) was involved in neuroprotective effects of APX3330 in enteric neurons. CONCLUSIONS This study is the first to investigate inhibition of APE1/Ref-1's redox activity via APX3330 in an animal model of chronic intestinal inflammation. Inhibition of the redox function of APE1/Ref-1 is a novel strategy that might lead to a possible application of APX3330 for the treatment of IBD.
Collapse
Affiliation(s)
- Lauren Sahakian
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Rhiannon T Filippone
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Rhian Stavely
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia,Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ainsley M Robinson
- Institute for Health and Sport, Victoria University; Western Centre for Health, Research and Education, Sunshine Hospital, Melbourne, Victoria, Australia
| | - Xu Sean Yan
- 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 y Unidad Asociada al Instituto de Química Médica (IQM) del Consejo Superior de Investigaciones Científicas (CSIC), Universidad Rey Juan Carlos (URJC), Alcorcón, Madrid, Spain,High Performance Research Group in Physiopathology and Pharmacology of the Digestive System at URJC, Alcorcón, Madrid, Spain
| | - Rajaraman Eri
- University of Tasmania, School of Health Sciences, Launceston, Tasmania, Australia
| | - Joel C Bornstein
- Department of Physiology, Melbourne University, Melbourne, Australia
| | - Mark R Kelley
- Indiana University Simon Comprehensive Cancer Center, Departments of Pediatrics, Biochemistry & Molecular Biology and Pharmacology & Toxicology, Program in Pediatric Molecular Oncology & Experimental Therapeutics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine Indianapolis, USA
| | - 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 of Musculoskeletal Science (AIMSS), Melbourne, Victoria, Australia,Address correspondence to: Kulmira Nurgali, Level 4, Research Labs, Western Centre for Health Research & Education, Sunshine Hospital, 176 Furlong Road, St Albans, 3021, VIC, Australia. E-mail:
| |
Collapse
|
21
|
Nyavor Y, Brands CR, May G, Kuther S, Nicholson J, Tiger K, Tesnohlidek A, Yasuda A, Starks K, Litvinenko D, Linden DR, Bhattarai Y, Kashyap PC, Forney LJ, Balemba OB. High-fat diet-induced alterations to gut microbiota and gut-derived lipoteichoic acid contributes to the development of enteric neuropathy. Neurogastroenterol Motil 2020; 32:e13838. [PMID: 32168415 PMCID: PMC7319907 DOI: 10.1111/nmo.13838] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/28/2020] [Accepted: 02/21/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND High-fat diet, microbial alterations and lipopolysaccharide (LPS) are thought to cause enteric diabetic neuropathy and intestinal dysmotility. However, the role of the gut microbiota, lipoteichoic acid (LTA) from Gram-positive bacteria and short-chain fatty acids (SCFAs) in the development of diabetic enteric neuropathy and intestinal dysmotility is not well understood. Our aim was to examine the role of the gut microbiota, LTA and SCFAs in the development of diabetic enteric neuropathy and intestinal dysmotility. METHODS We fed germ-free (GF) and conventionally raised (CR) mice either a high-fat (HFD) or standard chow diet (SCD) for 8 weeks. We analyzed the microbial community composition in CR mice using 16S rRNA sequencing and damage to myenteric neurons using immunohistochemistry. We also studied the effects of LPS, LTA, and SCFAs on duodenal muscularis externa contractions and myenteric neurons using cultured preparations. KEY RESULTS High-fat diet ingestion reduced the total number and the number of nitrergic myenteric neurons per ganglion in the duodenum of CR but not in GF-HFD mice. GF mice had fewer neurons per ganglion compared with CR mice. CR mice fed a HFD had increased abundance of Gram-positive bacteria. LTA and LPS did not affect the frequency of duodenal muscularis contractions after 24 hours of cultured but reduced the density of nitrergic myenteric neurons and increased oxidative stress and TNFα production in myenteric ganglia. SCFAs did not affect muscularis contractions or injure myenteric neurons. CONCLUSIONS & INFERENCES Gut microbial alterations induced increase in Gram-positive bacterial LTA may contribute to enteric neuropathy.
Collapse
Affiliation(s)
- Yvonne Nyavor
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | | | - George May
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | - Sydney Kuther
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | | | - Kathryn Tiger
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | | | - Allysha Yasuda
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | - Kiefer Starks
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | - Diana Litvinenko
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | - David R. Linden
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Yogesh Bhattarai
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Purna C. Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Larry J. Forney
- University of Idaho, 875 Perimeter Drive, LSS 252 Moscow, ID 83844
| | | |
Collapse
|
22
|
Kong W, Li C, Qi Q, Shen J, Chang K. Cardamonin induces G2/M arrest and apoptosis via activation of the JNK-FOXO3a pathway in breast cancer cells. Cell Biol Int 2020; 44:177-188. [PMID: 31393045 DOI: 10.1002/cbin.11217] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/04/2019] [Indexed: 01/24/2023]
Abstract
Cardamonin (CD), a naturally occurring chalcone isolated from large black cardamom, was previously reported to suppress the proliferation of breast cancer cells. However, its precise molecular anti-tumor mechanisms have not been well elucidated. In this study, we found that CD markedly inhibited the proliferation of MDA-MB 231 and MCF-7 breast cancer cells through the induction of G2/M arrest and apoptosis. Reactive oxygen species (ROS) plays a pivotal role in the inhibition of CD-induced cell proliferation. Treatment with N-acetyl-cysteine (NAC), an ROS scavenger, blocked CD-induced G2/M arrest and apoptosis in this study. Quenching of ROS by overexpression of catalase also blocked CD-induced cell cycle arrest and apoptosis. We showed that CD enhanced the expression and nuclear translocation of Forkhead box O3 (FOXO3a) via upstream c-Jun N-terminal kinase, inducing the expression of FOXO3a and its target genes, including p21, p27, and Bim. This process led to the reduction of cyclin D1 and enhancement of activated caspase-3 expression. The addition of NAC markedly reversed these effects, knockdown of FOXO3a using small interfering RNA also decreased CD-induced G2/M arrest and apoptosis. In vivo, CD efficiently suppressed the growth of MDA-MB 231 breast cancer xenograft tumors. Taken together, our data provide a molecular mechanistic rationale for CD-induced cell cycle arrest and apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Weiwei Kong
- Department of Blood Transfusion, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, China
| | - Chuang Li
- Department of Laboratory Medicine, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, China
| | - Qiaofang Qi
- Xinxiang Medical University, 453000, Xinxiang, China
| | - Jiahui Shen
- Department of Blood Transfusion, The Third Affiliated Hospital of Xinxiang Medical University, 453000, Xinxiang, China
| | - Kaiwen Chang
- Xinxiang Medical University, 453000, Xinxiang, China
| |
Collapse
|
23
|
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: 3.7] [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
|
24
|
Boesmans W, Hao MM, Fung C, Li Z, Van den Haute C, Tack J, Pachnis V, Vanden Berghe P. Structurally defined signaling in neuro-glia units in the enteric nervous system. Glia 2019; 67:1167-1178. [PMID: 30730592 PMCID: PMC6593736 DOI: 10.1002/glia.23596] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/11/2019] [Accepted: 01/11/2019] [Indexed: 12/21/2022]
Abstract
Coordination of gastrointestinal function relies on joint efforts of enteric neurons and glia, whose crosstalk is vital for the integration of their activity. To investigate the signaling mechanisms and to delineate the spatial aspects of enteric neuron-to-glia communication within enteric ganglia we developed a method to stimulate single enteric neurons while monitoring the activity of neighboring enteric glial cells. We combined cytosolic calcium uncaging of individual enteric neurons with calcium imaging of enteric glial cells expressing a genetically encoded calcium indicator and demonstrate that enteric neurons signal to enteric glial cells through pannexins using paracrine purinergic pathways. Sparse labeling of enteric neurons and high-resolution analysis of the structural relation between neuronal cell bodies, varicose release sites and enteric glia uncovered that this form of neuron-to-glia communication is contained between the cell body of an enteric neuron and its surrounding enteric glial cells. Our results reveal the spatial and functional foundation of neuro-glia units as an operational cellular assembly in the enteric nervous system.
Collapse
Affiliation(s)
- Werend Boesmans
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.,Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.,Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Marlene M Hao
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Australia
| | - Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Zhiling Li
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, University of Leuven, Leuven, Belgium.,Leuven Viral Vector Core, University of Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Vassilis Pachnis
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Morales-Soto W, Gulbransen BD. Enteric Glia: A New Player in Abdominal Pain. Cell Mol Gastroenterol Hepatol 2018; 7:433-445. [PMID: 30739868 PMCID: PMC6369218 DOI: 10.1016/j.jcmgh.2018.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Chronic abdominal pain is the most common gastrointestinal issue and contributes to the pathophysiology of functional bowel disorders and inflammatory bowel disease. Current theories suggest that neuronal plasticity and broad alterations along the brain-gut axis contribute to the development of chronic abdominal pain, but the specific mechanisms involved in chronic abdominal pain remain incompletely understood. Accumulating evidence implicates glial cells in the development and maintenance of chronic pain. Astrocytes and microglia in the central nervous system and satellite glia in dorsal root ganglia contribute to chronic pain states through reactive gliosis, the modification of glial networks, and the synthesis and release of neuromodulators. In addition, new data suggest that enteric glia, a unique type of peripheral glia found within the enteric nervous system, have the potential to modify visceral perception through interactions with neurons and immune cells. Understanding these emerging roles of enteric glia is important to fully understand the mechanisms that drive chronic pain and to identify novel therapeutic targets. In this review, we discuss enteric glial cell signaling mechanisms that have the potential to influence chronic abdominal pain.
Collapse
Affiliation(s)
| | - Brian D. Gulbransen
- Correspondence Address correspondence to: Brian D. Gulbransen, PhD, Neuroscience Program and Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, Michigan 48824. fax: (517) 355-5125.
| |
Collapse
|
26
|
Rytel L, Szymanska K, Gonkowski I, Wojtkiewicz J. Neurochemical characterization of intramural nerve fibres in the porcine oesophagus. Anat Histol Embryol 2018; 47:517-526. [PMID: 30105873 DOI: 10.1111/ahe.12391] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/14/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
The gastrointestinal (GI) tract is innervated by nerve processes derived from the intramural enteric neurons and neurons localized outside the digestive tract. This study analysed the neurochemical characterization of nerves in the wall of the porcine oesophagus using single immunofluorescence technique. Immunoreactivity to vesicular acetylcholine transporter (VAChT), neuropeptide Y (NPY), vasoactive intestinal polypeptide (VIP), somatostatin (SOM), galanin (GAL), neuronal isoform of nitric oxide synthase (nNOS), substance P (SP), leucine enkephalin (LENK), calcitonin gene-related peptide (CGRP) or dopamine beta-hydroxylase (DBH) was investigated in intramuscular and intramucosal nerves of the cervical, thoracic and abdominal oesophagus. The results indicate that all of the substances studied were present in the oesophageal nerves. The density of particular populations of fibres depended on the segment of the oesophagus. The most numerous were fibres immunoreactive to VIP in the longitudinal and circular muscle layers of the abdominal oesophagus: The number of these fibres amounted to 16.4 ± 0.8 and 18.1 ± 3.1, respectively. In turn, the least numerous were CGRP-positive fibres, which were present only in the circular muscle layer of the cervical oesophagus and mucosal layer of the abdominal oesophagus in the number of 0.3 ± 0. The obtained results show that nerves in the porcine oesophageal wall are very diverse in their neurochemical coding, and differences between particular parts of the oesophagus suggest that organization of the innervation clearly depends on the fragment of this organ.
Collapse
Affiliation(s)
- Liliana Rytel
- Faculty of Veterinary Medicine, Department of Internal Disease with Clinic, University of Warmia and Mazury, Olsztyn, Poland
| | - Kamila Szymanska
- Faculty of Veterinary Medicine, Department of Clinical Physiology, University of Warmia and Mazury, Olsztyn, Poland
| | - Ignacy Gonkowski
- Department of Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| |
Collapse
|