1
|
O'Riordan KJ, Moloney GM, Keane L, Clarke G, Cryan JF. The gut microbiota-immune-brain axis: Therapeutic implications. Cell Rep Med 2025; 6:101982. [PMID: 40054458 DOI: 10.1016/j.xcrm.2025.101982] [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: 05/24/2024] [Revised: 10/15/2024] [Accepted: 01/29/2025] [Indexed: 03/21/2025]
Abstract
The microbiota-gut-brain axis has major implications for human health including gastrointestinal physiology, brain function, and behavior. The immune system represents a key pathway of communication along this axis with the microbiome implicated in neuroinflammation in health and disease. In this review, we discuss the mechanisms as to how the gut microbiota interacts with the brain, focusing on innate and adaptive immunity that are often disrupted in gut-brain axis disorders. We also consider the implications of these observations and how they can be advanced by interdisciplinary research. Leveraging an increased understanding of how these interactions regulate immunity has the potential to usher in a new era of precision neuropsychiatric clinical interventions for psychiatric, neurodevelopmental, and neurological disorders.
Collapse
Affiliation(s)
| | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Lily Keane
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, T12 YT20 Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
2
|
Costa CJ, Prescott S, Fourie NH, Abey SK, Sherwin LB, Rahim-Williams B, Joseph PV, Posada-Quintero H, Hoffman RK, Henderson WA. Host Transcriptome and Microbial Variation in Relation to Visceral Hyperalgesia. Nutrients 2025; 17:921. [PMID: 40077792 PMCID: PMC11902232 DOI: 10.3390/nu17050921] [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: 02/07/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Chronic visceral hypersensitivity is associated with an overstressed pain response to noxious stimuli (hyperalgesia). Microbiota are active modulators of host biology and are implicated in the etiology of visceral hypersensitivity. OBJECTIVES we studied the association between the circulating mRNA transcriptome, the intensity of induced visceral pain (IVP), and variation in the oral microbiome among participants with and without baseline visceral hypersensitivity. METHODS Transcriptomic profiles and microbial abundance were correlated with IVP intensity. Host mRNA and microbes associated with IVP were explored, linking variation in the microbiome to host RNA biology. RESULTS 259 OTUs were found to be associated with IVP through correlation to differential expression of 471 genes in molecular pathways related to inflammation and neural mechanisms, including Rho and PI3K/AKT pathways. The bacterial families Lachnospiraceae, Prevotellaceae, and Veillonellaceae showed the highest degree of association. Oral microbial profiles with reduced diversity were characteristic of participants with visceral hypersensitivity. CONCLUSIONS Our results suggest that the oral microbiome may be involved in systemic immune and inflammatory effects and play a role in nervous system and stem cell pathways. The interactions between visceral hypersensitivity, differentially expressed molecular pathways, and microbiota described here provide a framework for further work exploring the relationship between host and microbiome.
Collapse
Affiliation(s)
- Christopher J. Costa
- Department of Medicine, UConn Health, 263 Farmington Ave, Farmington, CT 06030, USA;
| | - Stephanie Prescott
- Inova Health Services, L.J. Murphy Children’s Hospital, 3300 Gallows Rd, Falls Church, VA 22042, USA;
| | - Nicolaas H. Fourie
- National Institute of Nursing Research, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, USA
| | - Sarah K. Abey
- Laboratory of Neuroimaging, National Institute of Alcohol Abuse and Alcoholism, 10 Center Drive, Bethesda, MD 20814, USA
| | - LeeAnne B. Sherwin
- Sinclair School of Nursing, University of Missouri System, 915 Hitt Street, Columbia, MO 65203, USA;
| | - Bridgett Rahim-Williams
- Office of Research and Sponsored Programs, University of North Florida, 1 UNF Drive, Jacksonville, FL 32224, USA;
| | - Paule V. Joseph
- National Institute on Alcohol Abuse and Alcoholism, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 10 Center Drive, Bethesda, MD 20814, USA;
| | - Hugo Posada-Quintero
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Storrs, CT 06269, USA;
| | - Rebecca K. Hoffman
- Laboratory of Innovative and Translational Nursing Research, School of Nursing, University of PA, 418 Curie Blvd, Philadelphia, PA 19104, USA;
| | - Wendy A. Henderson
- Department of Biobehavioral Health Sciences, School of Nursing, University of PA, 418 Curie Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Mallick K, Khodve G, Ruwatia R, Banerjee S. Gut microbes: Therapeutic Target for neuropsychiatric disorders. J Psychiatr Res 2025; 184:27-38. [PMID: 40036939 DOI: 10.1016/j.jpsychires.2025.02.031] [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: 11/01/2024] [Revised: 01/28/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Neuropsychiatric diseases encompass a range of mental and neurological disorders that have a significant and far-reaching effect on an individual's quality of life. These conditions affect not only the mental status but also the physical well-being of individuals, which leads to weakened immune systems and other diseases. Emerging research underscores a significant connection between the gut microbiome and neuropsychiatric diseases, suggesting that microbial communities within the gastrointestinal tract may influence brain function and mental health. Gut dysbiosis is caused by various factors, including stress, diet, inappropriate usage of antibiotics, infections, and so on, all of which can disrupt numerous pathways, resulting in abnormal neurotransmitter signaling, inflammation, and impaired brain function. Similarly, various neuropsychiatric diseases can disrupt the specific microbiome in the gut, leading to gut dysbiosis, often impairing memory and cognitive function. The growing evidence supporting the role of gut dysbiosis in neuropsychiatric disorders has opened up new avenues for therapeutic interventions. Modulating the gut microbiome through strategies such as probiotics, prebiotics, or fecal microbiota transplantation has shown promising results in various studies of neuropsychiatric disorders. However, further research is needed to fully elucidate the mechanisms involved in gut dysbiosis-associated brain changes to develop effective and personalized treatment strategies for neuropsychiatric diseases.
Collapse
Affiliation(s)
- Keya Mallick
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Gopal Khodve
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Ritika Ruwatia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
4
|
Bellés A, Abad I, Buey B, Vergara C, Mesonero JE, Sánchez L, Grasa L. Buttermilk and Whey as Functional Foods to Ameliorate Clindamycin-Induced Changes in Mouse Intestine: Modulation of Intestinal Motility and Toll-like Receptors Expression. J Med Food 2025; 28:205-211. [PMID: 39509172 DOI: 10.1089/jmf.2024.0155] [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: 11/15/2024] Open
Abstract
Antibiotic treatment is one of the main causes of intestinal dysbiosis, leading, in turn, to other intestinal alterations given the multiple relationships of the microbiota with gut health. Whey and buttermilk are two by-products from the dairy industry with numerous bioactive components. This study aimed to assess the potential of two formulas, containing a mixture of lactoferrin, milk fat globule membrane (MFGM), and whey or buttermilk, to reverse the negative effects of clindamycin on gut motility, Toll-like receptors (TLRs) expression, and oxidative stress in the intestine. For this purpose, a murine model of intestinal dysbiosis was established by clindamycin treatment. Male C57BL/6 mice were treated with saline (Control), clindamycin (Clin), a formula containing whey (F1), or buttermilk (F2) supplemented with lactoferrin and MFGM, Clin+F1, or Clin+F2. Clin delayed the whole gut transit, reduced the response to acetylcholine, decreased TLR2 expression, and increased TLR4 expression in the intestine. F1 and F2 formulas reversed the effects of Clin, restoring TLR2 receptor levels and normalizing intestinal dysmotility. These results indicate that whey- and buttermilk-based formulas supplemented with lactoferrin and MFGM could be used as functional foods to prevent or treat motility disorders and restore some components of the immune system after antibiotic treatment.
Collapse
Affiliation(s)
- Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense. Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), Zaragoza, Spain
| | - Inés Abad
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), Zaragoza, Spain
- Departamento de Producción Animal y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Berta Buey
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense. Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - Claudia Vergara
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense. Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - José Emilio Mesonero
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense. Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - Lourdes Sánchez
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), Zaragoza, Spain
- Departamento de Producción Animal y Tecnología de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense. Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| |
Collapse
|
5
|
Ramírez-Maldonado LM, Guerrero-Castro J, Rodríguez-Mejía JL, Cárdenas-Conejo Y, Bonales-Alatorre EO, Valencia-Cruz G, Anguiano-García PT, Vega-Juárez II, Dagnino-Acosta A, Ruvalcaba-Galindo J, Valdez-Morales EE, Ochoa-Cortes F, Barajas-Espinosa A, Guerrero-Alba R, Liñán-Rico A. Obesogenic cafeteria diet induces dynamic changes in gut microbiota, reduces myenteric neuron excitability, and impairs gut contraction in mice. Am J Physiol Gastrointest Liver Physiol 2025; 328:G32-G48. [PMID: 39499253 DOI: 10.1152/ajpgi.00198.2024] [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: 07/02/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/07/2024]
Abstract
The cafeteria diet (CAF) is a superior diet model in animal experiments compared with the conventional high-fat diet (HFD), effectively inducing obesity, metabolic disturbances, and multi-organ damage. Nevertheless, its impact on gut microbiota composition during the progression of obesity, along with its repercussions on the enteric nervous system (ENS) and gastrointestinal motility has not been completely elucidated. To gain more insight into the effects of CAF diet in the gut, C57BL/6 mice were fed with CAF or a standard diet for 2 or 8 wk. CAF-fed mice experienced weight gain, disturbed glucose metabolism, dysregulated expression of colonic IL-6, IL-22, TNFα, and TPH1, and altered colon morphology, starting at week 2. Fecal DNA was isolated and gut microbiota composition was monitored by sequencing the V3-V4 16S rRNA region. Sequence analysis revealed that Clostridia and Proteobacteria were specific biomarkers associated with CAF-feeding at week 2, while Bacteroides and Actinobacteria were prominent at week 8. In addition, the impact of CAF diet on ENS was investigated (week 8), where HuC/D+ neurons were measured and counted, and their biophysical properties were evaluated by patch clamp. Gut contractility was tested in whole-mount preparations. Myenteric neurons in CAF-fed mice exhibited reduced body size, incremented cell density, and decreased excitability. The amplitude and frequency of the rhythmic spontaneous contractions in the colon and ileum were affected by the CAF diet. Our findings demonstrate, for the first time, that CAF diet gradually changes the gut microbiota and promotes low-grade inflammation, impacting the functional properties of myenteric neurons and gut contractility in mice.NEW & NOTEWORTHY The gut microbiota changes gradually following the consumption of CAF diet. An increase in Clostridia and Proteobacteria is a hallmark of dysbiosis at the early onset of gut inflammation and obesity. The CAF diet was effective in inducing intestinal low-grade inflammation and alterations in myenteric neuronal excitability in mice. CAF diet is a reliable strategy to study the interplay between gut dysbiosis and low-grade inflammation, in addition to the mechanisms underlying gastrointestinal dysmotility associated with obesity.
Collapse
Affiliation(s)
- Luis M Ramírez-Maldonado
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
| | - Julio Guerrero-Castro
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
| | - José L Rodríguez-Mejía
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
| | - Yair Cárdenas-Conejo
- Laboratorio de Biología Sintética Estructural y Molecular, Universidad de Colima-Consejo Nacional de Humanidades, Ciencias y Tecnologías, Colima, México
| | - Edgar O Bonales-Alatorre
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
| | - Georgina Valencia-Cruz
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
| | | | - Irving I Vega-Juárez
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
| | - Adán Dagnino-Acosta
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima-Consejo Nacional de Humanidades, Ciencias y Tecnologías, Colima, México
| | | | - Eduardo E Valdez-Morales
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes-Consejo Nacional de Humanidades, Ciencias y Tecnologías, Aguascalientes, México
| | - Fernando Ochoa-Cortes
- Escuela Superior de Huejutla, Universidad Autónoma del Estado de Hidalgo, Huejutla de Reyes, Hidalgo, México
| | - Alma Barajas-Espinosa
- Escuela Superior de Huejutla, Universidad Autónoma del Estado de Hidalgo, Huejutla de Reyes, Hidalgo, México
| | - Raquel Guerrero-Alba
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, México
| | - Andrómeda Liñán-Rico
- Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, México
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima-Consejo Nacional de Humanidades, Ciencias y Tecnologías, Colima, México
| |
Collapse
|
6
|
Ghosh A, Gorain B. Mechanistic insight of neurodegeneration due to micro/nano-plastic-induced gut dysbiosis. Arch Toxicol 2025; 99:83-101. [PMID: 39370473 DOI: 10.1007/s00204-024-03875-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Despite offering significant conveniences, plastic materials contribute substantially in developing environmental hazards and pollutants. Plastic trash that has not been adequately managed may eventually break down into fragments caused by human or ecological factors. Arguably, the crucial element for determining the biological toxicities of plastics are micro/nano-forms of plastics (MPs/NPs), which infiltrate the mammalian tissue through different media and routes. Infiltration of MPs/NPs across the intestinal barrier leads to microbial architectural dysfunction, which further modulates the population of gastrointestinal microbes. Thereby, it triggers inflammatory mediators (e.g., IL-1α/β, TNF-α, and IFN-γ) by activating specific receptors located in the gut barrier. Mounting evidence indicates that MPs/NPs disrupt host pathophysiological function through modification of junctional proteins and effector cells. Moreover, the alteration of microbial diversity by MPs/NPs causes the breakdown of the blood-brain barrier and translocation of metabolites (e.g., SCFAs, LPS) through the vagus nerve. Potent penetration affects the neuronal networks, neuronal protein accumulation, acceleration of oxidative stress, and alteration of neurofibrillary tangles, and hinders distinctive communicating pathways. Conclusively, alterations of these neurotoxic factors are possibly responsible for the associated neurodegenerative disorders due to the exposure of MPs/NPs. In this review, the hypothesis on MPs/NPs associated with gut microbial dysbiosis has been interlinked to the distinct neurological impairment through the gut-brain axis.
Collapse
Affiliation(s)
- Arya Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Bapi Gorain
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| |
Collapse
|
7
|
Hong SM, Qian X, Deshpande V, Kulkarni S. Optimization of protocols for immunohistochemical assessment of enteric nervous system in formalin fixed human tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.15.628584. [PMID: 39763767 PMCID: PMC11702535 DOI: 10.1101/2024.12.15.628584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Gastrointestinal (GI) motility is regulated in a large part by the cells of the enteric nervous system (ENS), suggesting that ENS dysfunctions either associate with, or drive GI dysmotility in patients. However, except for select diseases such as Hirschsprung's Disease or Achalasia that show a significant loss of all neurons or a subset of neurons, our understanding of human ENS histopathology is extremely limited. Recent endoscopic advances allow biopsying patient's full thickness gut tissues, which makes capturing ENS tissues simpler than biopsying other neuronal tissues, such as the brain. Yet, our understanding of ENS aberrations observed in GI dysmotility patients lags behind our understanding of central nervous system aberrations observed in patients with neurological disease. Paucity of optimized methods for histopathological assessment of ENS in pathological specimens represent an important bottleneck in ascertaining how the ENS is altered in diverse GI dysmotility conditions. While recent studies have interrogated ENS structure in surgically resected whole mount human gut, most pathological specimens are banked as formalin fixed paraffin embedded (FFPE) tissue blocks - suggesting that methods to interrogate ENS in FFPE tissue blocks would provide the biggest impetus for ENS histopathology in a clinical setting. In this report, we present optimized methods for immunohistochemical interrogation of the human ENS tissue on the basis of >25 important protein markers that include proteins expressed by all neurons, subset of neurons, hormones, and neurotransmitter receptors. This report provides a resource which will help pathologists and investigators assess ENS aberrations in patients with various GI dysmotility conditions.
Collapse
Affiliation(s)
- Su Min Hong
- Division of Gastroenterology, Dept of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Xia Qian
- Dept of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Vikram Deshpande
- Dept of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115
| | - Subhash Kulkarni
- Division of Gastroenterology, Dept of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115
- Division of Medical Sciences, Harvard Medical School, Boston, MA 02115
- Graduate program in Neuroscience, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
8
|
Pan I, Issac PK, Rahman MM, Guru A, Arockiaraj J. Gut-Brain Axis a Key Player to Control Gut Dysbiosis in Neurological Diseases. Mol Neurobiol 2024; 61:9873-9891. [PMID: 37851313 DOI: 10.1007/s12035-023-03691-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/02/2023] [Indexed: 10/19/2023]
Abstract
Parkinson's disease is a chronic neuropathy characterised by the formation of Lewy bodies (misfolded alpha-synuclein) in dopaminergic neurons of the substantia nigra and other parts of the brain. Dopaminergic neurons play a vital role in generating both motor and non-motor symptoms. Finding therapeutic targets for Parkinson's disease (PD) is hindered due to an incomplete understanding of the disease's pathophysiology. Existing evidence suggests that the gut microbiota participates in the pathogenesis of PD via immunological, neuroendocrine, and direct neural mechanisms. Gut microbial dysbiosis triggers the loss of dopaminergic neurons via mitochondrial dysfunction. Gut dysbiosis triggers bacterial overgrowth in the small intestine, which increases the permeability barrier and induces systemic inflammation. It results in excessive stimulation of the innate immune system. In addition to that, activation of enteric neurons and enteric glial cells initiates the aggregation of alpha-synuclein. This alpha-synucleinopathy thus affects all levels of the brain-gut axis, including the central, autonomic, and enteric nervous systems. Though the neurobiological signaling cascade between the gut microbiome and the central nervous system is poorly understood, gut microbial metabolites may serve as a promising therapeutic strategy for PD. This article summarises all the known possible ways of bidirectional signal communication, i.e., the "gut-brain axis," where microbes from the middle gut interact with the brain and vice versa, and highlights a unique way to treat neurodegenerative diseases by maintaining homeostasis. The tenth cranial nerve (vagus nerve) plays a significant part in this signal communication. However, the leading regulatory factor for this axis is a diet that helps with microbial colonisation and brain function. Short-chain fatty acids (SCFAs), derived from microbially fermented dietary fibres, link host nutrition to maintain intestinal homeostasis. In addition to that, probiotics modulate cognitive function and the metabolic and behavioural conditions of the body. As technology advances, new techniques will emerge to study the tie-up between gut microbes and neuronal diseases.
Collapse
Affiliation(s)
- Ieshita Pan
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, Tamil Nadu, 602105, India.
| | - Praveen Kumar Issac
- Institute of Biotechnology, Department of Medical Biotechnology and Integrative Physiology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, Tamil Nadu, 602105, India
| | - Md Mostafizur Rahman
- Laboratory of Environmental Health and Ecotoxicology, Department of Environmental Sciences, Jahangirnagar University, Dhaka, 1342, Bangladesh
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulatur, Chengalpattu District, Tamil Nadu, 603203, India.
| |
Collapse
|
9
|
Kato R, Yamamoto T, Ogata H, Miyata K, Hayashi S, Gershon MD, Kadowaki M. Indigenous gut microbiota constitutively drive release of ciliary neurotrophic factor from mucosal enteric glia to maintain the homeostasis of enteric neural circuits. Front Immunol 2024; 15:1372670. [PMID: 39606241 PMCID: PMC11598343 DOI: 10.3389/fimmu.2024.1372670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/09/2024] [Indexed: 11/29/2024] Open
Abstract
It has recently become clear that the gut microbiota influence intestinal motility, intestinal barrier function, and mucosal immune function; therefore, the gut microbiota are deeply involved in the maintenance of intestinal homeostasis. The effects of the gut microbiota on the enteric nervous system (ENS) in the adult intestine, however, remain poorly understood. In the current study, we investigated the effects of the gut microbiota on the ENS. Male C57BL/6 SPF mice at 12 weeks of age were given a cocktail of four antibiotics (ABX) orally to induce dysbiosis (ABX mice). As early as six hours after ABX administration, the weight of the cecum of ABX mice increased to be significantly greater than that of vehicle-treated animals; moreover, ABX-induced dysbiosis reduced the density of enteric nerve fibers (marked by tubulin-β3 immunoreactivity) in the lamina propria of the proximal colon to approximately 60% that of control. TAK242, a TLR4 antagonist, significantly lowered the nerve fiber density in the lamina propria of the proximal colonic mucosa to approximately 60% that of vehicle-treated SPF mice. We thus developed and tested the hypothesis that mucosal glia expressing TLR4 are activated by enteric bacteria and release neurotrophic factors that contribute to the maintenance of enteric neural circuits. Neurotrophic factors in the mucosa of the SPF mouse proximal colon were examined immunohistochemically. Ciliary neurotrophic factor (CNTF) was abundantly expressed in the lamina propria; most of the CNTF immunoreactivity was observed in mucosal glia (marked by S100β immunoreactivity). Administration of CNTF (subcutaneously, 0.3 mg/kg, 3 doses, 2 hours apart) to ABX mice significantly increased mucosal nerve fiber density in the ABX mouse proximal colon to nearly control levels. The effect of CNTF on enteric mucosal nerve fibers was examined in isolated preparations of proximal colon of ABX mice. As it did in vivo, exposure to CNTF in vitro significantly increased enteric mucosal nerve fiber density in the ABX-treated colon. In conclusion, our evidence suggests that gut microbiota constitutively activate TLR4 signaling in enteric mucosal glia, which secrete CNTF in response. The resulting bacterial-driven glial release of CNTF helps to maintain the integrity of enteric mucosal nerve fibers.
Collapse
Affiliation(s)
- Ryo Kato
- Division of Gastrointestinal Pathophysiology, University of Toyama, Toyama, Japan
| | - Takeshi Yamamoto
- Division of Gastrointestinal Pathophysiology, University of Toyama, Toyama, Japan
| | - Hanako Ogata
- Division of Gastrointestinal Pathophysiology, University of Toyama, Toyama, Japan
| | - Kana Miyata
- Division of Gastrointestinal Pathophysiology, University of Toyama, Toyama, Japan
| | - Shusaku Hayashi
- Division of Gastrointestinal Pathophysiology, University of Toyama, Toyama, Japan
| | - Michael D. Gershon
- Departments of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | - Makoto Kadowaki
- Division of Gastrointestinal Pathophysiology, University of Toyama, Toyama, Japan
| |
Collapse
|
10
|
Lu S, Zhao Q, Guan Y, Sun Z, Li W, Guo S, Zhang A. The communication mechanism of the gut-brain axis and its effect on central nervous system diseases: A systematic review. Biomed Pharmacother 2024; 178:117207. [PMID: 39067168 DOI: 10.1016/j.biopha.2024.117207] [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: 05/13/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Gut microbiota is involved in intricate and active metabolic processes the host's brain function, especially its role in immune responses, secondary metabolism, and symbiotic connections with the host. Gut microbiota can promote the production of essential metabolites, neurotransmitters, and other neuroactive chemicals that affect the development and treatment of central nervous system diseases. This article introduces the relevant pathways and manners of the communication between the brain and gut, summarizes a comprehensive overview of the current research status of key gut microbiota metabolites that affect the functions of the nervous system, revealing those adverse factors that affect typical communication between the brain-gut axis, and outlining the efforts made by researchers to alleviate these neurological diseases through targeted microbial interventions. The relevant pathways and manners of communication between the brain and gut contribute to the experimental design of new treatment plans and drug development. The factors that may cause changes in gut microbiota and affect metabolites, as well as current intervention methods are summarized, which helps improve gut microbiota brain dialogue, prevent adverse triggering factors from interfering with the gut microbiota system, and minimize neuropathological changes.
Collapse
Affiliation(s)
- Shengwen Lu
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Qiqi Zhao
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Yu Guan
- Department of Pharmaceutical Analysis, GAP Center, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Zhiwen Sun
- Department of Gastroenterology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Wenhao Li
- School of Basic Medical Science of Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China
| | - Sifan Guo
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China
| | - Aihua Zhang
- International Advanced Functional Omics Platform, Scientific Experiment Center, Hainan Medical University, Xueyuan Road 3, Haikou 571199, China; Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China; INTI International University, Nilai 71800, Malaysia.
| |
Collapse
|
11
|
Liu Y, Jia N, Tang C, Long H, Wang J. Microglia in Microbiota-Gut-Brain Axis: A Hub in Epilepsy. Mol Neurobiol 2024; 61:7109-7126. [PMID: 38366306 DOI: 10.1007/s12035-024-04022-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/06/2024] [Indexed: 02/18/2024]
Abstract
There is growing concern about the role of the microbiota-gut-brain axis in neurological illnesses, and it makes sense to consider microglia as a critical component of this axis in the context of epilepsy. Microglia, which reside in the central nervous system, are dynamic guardians that monitor brain homeostasis. Microglia receive information from the gut microbiota and function as hubs that may be involved in triggering epileptic seizures. Vagus nerve bridges the communication in the axis. Essential axis signaling molecules, such as gamma-aminobutyric acid, 5-hydroxytryptamin, and short-chain fatty acids, are currently under investigation for their participation in drug-resistant epilepsy (DRE). In this review, we explain how vagus nerve connects the gut microbiota to microglia in the brain and discuss the emerging concepts derived from this interaction. Understanding microbiota-gut-brain axis in epilepsy brings hope for DRE therapies. Future treatments can focus on the modulatory effect of the axis and target microglia in solving DRE.
Collapse
Affiliation(s)
- Yuyang Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Ningkang Jia
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
- The Second Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Chuqi Tang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China
| | - Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- The First Clinical Medicine College, Southern Medical University, Guangzhou, China.
- Neural Networks Surgery Team, Southern Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Zhang X, Chen J, He F, Du W, Yu X. Assessing the causal effects of Eubacterium and Rumphococcus on constipation: a Mendelian randomized study. Front Microbiol 2024; 15:1376232. [PMID: 39144218 PMCID: PMC11324052 DOI: 10.3389/fmicb.2024.1376232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Background Constipation is affected by a number of risk variables, including cardiovascular disease and growth factors. However, the impacts of gut flora on constipation incidence has not been shown. This work, Single-Variable Mendelian Randomization (SVMR) was utilized to estimate the causal relationship between the Eubacterium genus or Rumphococcus, and constipation. Methods Data for constipation, Eubacterium genus and Rumphococcus were taken from the Integrated Epidemiology Unit (IEU) open GWAS database. Including 218,792 constipation samples, and there were 16,380,466 Single Nucleotide Polymorphisms (SNPs) for constipation. The ids of Eubacterium genus and Rumphococcus were sourced from MiBioGen database. The sample count for the Eubacterium genus was 17,380, with 656 SNPs. In addition, the sample size for Rumphococcus was 15,339, with 545 SNPs. The SVMR was performed to assess the risk of Eubacterium genus and Rumphococcus in constipation using weighted median, MR Egger, simple mode, inverse variance weighted (IVW), and weighted mode. Finally, we did a sensitivity analysis that included a heterogeneity, horizontal pleiotropy, and Leave-One-Out (LOO) test to examine the viability of the MR data. Results The SVMR revealed that the Eubacterium genus and Rumphococcus were causally connected to constipation, with Rumphococcus (P = 0.042, OR = 1.074) as a hazardous factor and Eubacterium genus (P = 0.004, OR = 0.909) as a safety factor. Sensitivity tests then revealed the absence of variability between the constipation and the exposure factors (Eubacterium genus and Rumphococcus). Additionally, there were no other confounding factors and the examined SNPs could only influence constipation through the aforementioned exposure factors, respectively. As a result, the MR results were fairly robust. Conclusion Our investigation verified the causal links between the Eubacterium genus or Rumphococcus, and constipation, with greater Rumphococcus expression increasing the likelihood of constipation and the opposite being true for the Eubacterium genus.
Collapse
Affiliation(s)
- Xiao Zhang
- Guizhou University of Traditional Chinese Medicine, Department of Colorectal Medicine, Guiyang, Guizhou, China
| | - Jiang Chen
- Guizhou University of Traditional Chinese Medicine, Department of Colorectal Medicine, Guiyang, Guizhou, China
| | - Feng He
- Guizhou University of Traditional Chinese Medicine, Department of Colorectal Medicine, Guiyang, Guizhou, China
| | - Wenchun Du
- Guizhou University of Traditional Chinese Medicine, Department of Colorectal Medicine, Guiyang, Guizhou, China
| | - Xianhao Yu
- Guizhou University of Commerce, Computer and Information Engineering College, Guiyang, China
| |
Collapse
|
13
|
Cheong KL, Liu K, Chen W, Zhong S, Tan K. Recent progress in Porphyra haitanensis polysaccharides: Extraction, purification, structural insights, and their impact on gastrointestinal health and oxidative stress management. Food Chem X 2024; 22:101414. [PMID: 38711774 PMCID: PMC11070828 DOI: 10.1016/j.fochx.2024.101414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024] Open
Abstract
Porphyra haitanensis, a red seaweed species, represents a bountiful and sustainable marine resource. P. haitanensis polysaccharide (PHP), has garnered considerable attention for its numerous health benefits. However, the comprehensive utilization of PHP on an industrial scale has been limited by the lack of comprehensive information. In this review, we endeavor to discuss and summarize recent advancements in PHP extraction, purification, and characterization. We emphasize the multifaceted mechanisms through which PHP promotes gastrointestinal health. Furthermore, we present a summary of compelling evidence supporting PHP's protective role against oxidative stress. This includes its demonstrated potent antioxidant properties, its ability to neutralize free radicals, and its capacity to enhance the activity of antioxidant enzymes. The information presented here also lays the theoretical groundwork for future research into the structural and functional aspects of PHP, as well as its potential applications in functional foods.
Collapse
Affiliation(s)
- Kit-Leong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Keying Liu
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Wenting Chen
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Engineering Technology Research Center of Prefabricated Seafood Processing and Quality Control, Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Karsoon Tan
- Guangxi Key Laboratory of Beibu Gulf Biodiversity Conservation, Beibu Gulf University, Qinzhou, Guangxi, China
| |
Collapse
|
14
|
Gong Z, Xue Q, Luo Y, Yu B, Hua B, Liu Z. The interplay between the microbiota and opioid in the treatment of neuropathic pain. Front Microbiol 2024; 15:1390046. [PMID: 38919504 PMCID: PMC11197152 DOI: 10.3389/fmicb.2024.1390046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Neuropathic pain (NP) is characterized by its complex and multifactorial nature and limited responses to opioid therapy; NP is associated with risks of drug resistance, addiction, difficulty in treatment cessation, and psychological disorders. Emerging research on gut microbiota and their metabolites has demonstrated their effectiveness in alleviating NP and augmenting opioid-based pain management, concurrently mitigating the adverse effects of opioids. This review addresses the following key points: (1) the current advances in gut microbiota research and the challenges in using opioids to treat NP, (2) the reciprocal effects and benefits of gut microbiota on NP, and (3) the interaction between opioids with gut microbiota, as well as the benefits of gut microbiota in opioid-based treatment of NP. Through various intricate mechanisms, gut microbiota influences the onset and progression of NP, ultimately enhancing the efficacy of opioids in the management of NP. These insights pave the way for further pragmatic clinical research, ultimately enhancing the efficacy of opioid-based pain management.
Collapse
Affiliation(s)
- Zexiong Gong
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qingsheng Xue
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Buwei Yu
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Bo Hua
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhiheng Liu
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
15
|
Zhong D, Jin K, Wang R, Chen B, Zhang J, Ren C, Chen X, Lu J, Zhou M. Microalgae-Based Hydrogel for Inflammatory Bowel Disease and Its Associated Anxiety and Depression. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312275. [PMID: 38277492 DOI: 10.1002/adma.202312275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Indexed: 01/28/2024]
Abstract
Patients diagnosed with inflammatory bowel disease (IBD) exhibit a notable prevalence of psychiatric disorders, such as anxiety and depression. Nevertheless, the etiology of psychiatric disorders associated with IBD remains uncertain, and an efficacious treatment approach has yet to be established. Herein, an oral hydrogel strategy (SP@Rh-gel) is proposed for co-delivery of Spirulina platensis and rhein to treat IBD and IBD-associated anxiety and depression by modulating the microbiota-gut-brain axis. SP@Rh-gel improves the solubility, release characteristics and intestinal retention capacity of the drug, leading to a significant improvement in the oral therapeutic efficacy. Oral administration of SP@Rh-gel can reduce intestinal inflammation and rebalance the disrupted intestinal microbial community. Furthermore, SP@Rh-gel maintains intestinal barrier integrity and reduces the release of pro-inflammatory factors and their entry into the hippocampus through the blood-brain barrier, thereby inhibiting neuroinflammation and maintaining neuroplasticity. SP@Rh-gel significantly alleviates the colitis symptoms, as well as anxiety- and depression-like behaviors, in a chronic colitis mouse model. This study demonstrates the significant involvement of the microbiota-gut-brain axis in the development of IBD with psychiatric disorders and proposes a safe, simple, and highly efficient therapeutic approach for managing IBD and comorbid psychiatric disorders.
Collapse
Affiliation(s)
- Danni Zhong
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, P. R. China
| | - Kangyu Jin
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, P. R. China
| | - Ruoxi Wang
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, P. R. China
| | - Bing Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
| | - Jinghui Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, P. R. China
| | - Chaojie Ren
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Jing Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
- Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou, 310003, P. R. China
| | - Min Zhou
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, P. R. China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University, Haining, 314400, P. R. China
- National Key Laboratory of Biobased Transportation Fuel Technology, Zhejiang University, Hangzhou, 310027, P. R. China
- Zhejiang University-Erdos Etuoke Joint Research Center, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310029, P. R. China
| |
Collapse
|
16
|
Llorente C. The Imperative for Innovative Enteric Nervous System-Intestinal Organoid Co-Culture Models: Transforming GI Disease Modeling and Treatment. Cells 2024; 13:820. [PMID: 38786042 PMCID: PMC11119846 DOI: 10.3390/cells13100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.
Collapse
Affiliation(s)
- Cristina Llorente
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
17
|
Chen Y, Feng S, Li Y, Zhang C, Chao G, Zhang S. Gut microbiota and intestinal immunity-A crosstalk in irritable bowel syndrome. Immunology 2024; 172:1-20. [PMID: 38174581 DOI: 10.1111/imm.13749] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Irritable bowel syndrome (IBS), one of the most prevalent functional gastrointestinal disorders, is characterized by recurrent abdominal pain and abnormal defecation habits, resulting in a severe healthcare burden worldwide. The pathophysiological mechanisms of IBS are multi-factorially involved, including food antigens, visceral hypersensitivity reactions, and the brain-gut axis. Numerous studies have found that gut microbiota and intestinal mucosal immunity play an important role in the development of IBS in crosstalk with multiple mechanisms. Therefore, based on existing evidence, this paper elaborates that the damage and activation of intestinal mucosal immunity and the disturbance of gut microbiota are closely related to the progression of IBS. Combined with the application prospect, it also provides references for further in-depth exploration and clinical practice.
Collapse
Affiliation(s)
- Yuxuan Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyan Feng
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Li
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, China
| | - Guanqun Chao
- Department of General Practice, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou, China
| | - Shuo Zhang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
18
|
Sofield CE, Anderton RS, Gorecki AM. Mind over Microplastics: Exploring Microplastic-Induced Gut Disruption and Gut-Brain-Axis Consequences. Curr Issues Mol Biol 2024; 46:4186-4202. [PMID: 38785524 PMCID: PMC11120006 DOI: 10.3390/cimb46050256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
As environmental plastic waste degrades, it creates an abundance of diverse microplastic particles. Consequently, microplastics contaminate drinking water and many staple food products, meaning the oral ingestion of microplastics is an important exposure route for the human population. Microplastics have long been considered inert, however their ability to promote microbial dysbiosis as well as gut inflammation and dysfunction suggests they are more noxious than first thought. More alarmingly, there is evidence for microplastics permeating from the gut throughout the body, with adverse effects on the immune and nervous systems. Coupled with the now-accepted role of the gut-brain axis in neurodegeneration, these findings support the hypothesis that this ubiquitous environmental pollutant is contributing to the rising incidence of neurodegenerative diseases, like Alzheimer's disease and Parkinson's disease. This comprehensive narrative review explores the consequences of oral microplastic exposure on the gut-brain-axis by considering current evidence for gastrointestinal uptake and disruption, immune activation, translocation throughout the body, and neurological effects. As microplastics are now a permanent feature of the global environment, understanding their effects on the gut, brain, and whole body will facilitate critical further research and inform policy changes aimed at reducing any adverse consequences.
Collapse
Affiliation(s)
- Charlotte E. Sofield
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA 6160, Australia; (C.E.S.); (R.S.A.)
| | - Ryan S. Anderton
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA 6160, Australia; (C.E.S.); (R.S.A.)
- Institute for Health Research, University of Notre Dame Australia, Fremantle, WA 6160, Australia
| | - Anastazja M. Gorecki
- School of Health Sciences, University of Notre Dame Australia, Fremantle, WA 6160, Australia; (C.E.S.); (R.S.A.)
| |
Collapse
|
19
|
Thomasi B, Valdetaro L, Gulbransen B, Tavares-Gomes AL. Neuroimmune Connectomes in the Gut and Their Implications in Parkinson's Disease. Mol Neurobiol 2024; 61:2081-2098. [PMID: 37840070 PMCID: PMC11151216 DOI: 10.1007/s12035-023-03679-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
The gastrointestinal tract is the largest immune organ and it receives dense innervation from intrinsic (enteric) and extrinsic (sympathetic, parasympathetic, and somatosensory) neurons. The immune and neural systems of the gut communicate with each other and their interactions shape gut defensive mechanisms and neural-controlled gut functions such as motility and secretion. Changes in neuroimmune interactions play central roles in the pathogenesis of diseases such as Parkinson's disease (PD), which is a multicentric disorder that is heterogeneous in its manifestation and pathogenesis. Non-motor and premotor symptoms of PD are common in the gastrointestinal tract and the gut is considered a potential initiation site for PD in some cases. How the enteric nervous system and neuroimmune signaling contribute to PD disease progression is an emerging area of interest. This review focuses on intestinal neuroimmune loops such as the neuroepithelial unit, enteric glial cells and their immunomodulatory effects, anti-inflammatory cholinergic signaling and the relationship between myenteric neurons and muscularis macrophages, and the role of α-synuclein in gut immunity. Special consideration is given to the discussion of intestinal neuroimmune connectomes during PD and their possible implications for various aspects of the disease.
Collapse
Affiliation(s)
- Beatriz Thomasi
- Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building - Gulbransen lab, 567, Wilson Rd, Room 3199, East Lansing, MI, USA.
| | - Luisa Valdetaro
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, USA
| | - Brian Gulbransen
- Department of Physiology, Michigan State University, Biomedical and Physical Sciences Building - Gulbransen lab, 567, Wilson Rd, Room 3199, East Lansing, MI, USA
| | - Ana Lúcia Tavares-Gomes
- Programa de Pós-Graduação Em Neurociências, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Zhu J, He L. The Modulatory Effects of Curcumin on the Gut Microbiota: A Potential Strategy for Disease Treatment and Health Promotion. Microorganisms 2024; 12:642. [PMID: 38674587 PMCID: PMC11052165 DOI: 10.3390/microorganisms12040642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Curcumin (CUR) is a lipophilic natural polyphenol that can be isolated from the rhizome of turmeric. Studies have proposed that CUR possesses a variety of biological activities. Due to its anti-inflammatory and antioxidant properties, CUR shows promise in the treatment of inflammatory bowel disease, while its anti-obesity effects make it a potential therapeutic agent in the management of obesity. In addition, curcumin's ability to prevent atherosclerosis and its cardiovascular benefits further expand its potential application in the treatment of cardiovascular disease. Nevertheless, owing to the limited bioavailability of CUR, it is difficult to validate its specific mechanism of action in the treatment of diseases. However, the restricted bioavailability of CUR makes it challenging to confirm its precise mode of action in disease treatment. Recent research indicates that the oral intake of curcumin may lead to elevated levels of residual curcumin in the gastrointestinal system, hinting at curcumin's potential to directly influence gut microbiota. Furthermore, the ecological dysregulation of the gut microbiota has been shown to be critical in the pathogenesis of human diseases. This review summarizes the impact of gut dysbiosis on host health and the various ways in which curcumin modulates dysbiosis and ameliorates various diseases caused by it through the administration of curcumin.
Collapse
Affiliation(s)
- Junwen Zhu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China;
| | | |
Collapse
|
21
|
Liu W, Yan H, Jia W, Huang J, Fu Z, Xu W, Yu H, Yang W, Pan W, Zheng B, Liu Y, Chen X, Gao Y, Tian D. Association between gut microbiota and Hirschsprung disease: a bidirectional two-sample Mendelian randomization study. Front Microbiol 2024; 15:1366181. [PMID: 38516012 PMCID: PMC10956417 DOI: 10.3389/fmicb.2024.1366181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Background Several studies have pointed to the critical role of gut microbiota (GM) and their metabolites in Hirschsprung disease (HSCR) pathogenesis. However, the detailed causal relationship between GM and HSCR remains unknown. Methods In this study, we used two-sample Mendelian randomization (MR) analysis to investigate the causal relationship between GM and HSCR, based on the MiBioGen Consortium's genome-wide association study (GWAS) and the GWAS Catalog's HSCR data. Reverse MR analysis was performed subsequently, and the sensitivity analysis, Cochran's Q-test, MR pleiotropy residual sum, outlier (MR-PRESSO), and the MR-Egger intercept were used to analyze heterogeneity or horizontal pleiotropy. 16S rDNA sequencing and targeted mass spectrometry were developed for initial validation. Results In the forward MR analysis, inverse-variance weighted (IVW) estimates suggested that Eggerthella (OR: 2.66, 95%CI: 1.23-5.74, p = 0.01) was a risk factor for HSCR, while Peptococcus (OR: 0.37, 95%CI: 0.18-0.73, p = 0.004), Ruminococcus2 (OR: 0.32, 95%CI: 0.11-0.91, p = 0.03), Clostridiaceae1 (OR: 0.22, 95%CI: 0.06-0.78, p = 0.02), Mollicutes RF9 (OR: 0.27, 95%CI: 0.09-0.8, p = 0.02), Ruminococcaceae (OR: 0.16, 95%CI: 0.04-0.66, p = 0.01), and Paraprevotella (OR: 0.45, 95%CI: 0.21-0.98, p = 0.04) were protective factors for HSCR, which had no heterogeneity or horizontal pleiotropy. However, reverse MR analysis showed that HSCR (OR: 1.02, 95%CI: 1-1.03, p = 0.049) is the risk factor for Eggerthella. Furthermore, some of the above microbiota and short-chain fatty acids (SCFAs) were altered in HSCR, showing a correlation. Conclusion Our analysis established the relationship between specific GM and HSCR, identifying specific bacteria as protective or risk factors. Significant microbiota and SCFAs were altered in HSCR, underlining the importance of further study and providing new insights into the pathogenesis and treatment.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Hanlei Yan
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Wanying Jia
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Jingjing Huang
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Zihao Fu
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Wenyao Xu
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Hui Yu
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Weili Yang
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Weikang Pan
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Baijun Zheng
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yong Liu
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Xinlin Chen
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| | - Ya Gao
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Donghao Tian
- Department of Pediatric Surgery, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Chinese Ministry of Education, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Hung LY, Margolis KG. Autism spectrum disorders and the gastrointestinal tract: insights into mechanisms and clinical relevance. Nat Rev Gastroenterol Hepatol 2024; 21:142-163. [PMID: 38114585 DOI: 10.1038/s41575-023-00857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Autism spectrum disorders (ASDs) are recognized as central neurodevelopmental disorders diagnosed by impairments in social interactions, communication and repetitive behaviours. The recognition of ASD as a central nervous system (CNS)-mediated neurobehavioural disorder has led most of the research in ASD to be focused on the CNS. However, gastrointestinal function is also likely to be affected owing to the neural mechanistic nature of ASD and the nervous system in the gastrointestinal tract (enteric nervous system). Thus, it is unsurprising that gastrointestinal disorders, particularly constipation, diarrhoea and abdominal pain, are highly comorbid in individuals with ASD. Gastrointestinal problems have also been repeatedly associated with increased severity of the core symptoms diagnostic of ASD and other centrally mediated comorbid conditions, including psychiatric issues, irritability, rigid-compulsive behaviours and aggression. Despite the high prevalence of gastrointestinal dysfunction in ASD and its associated behavioural comorbidities, the specific links between these two conditions have not been clearly delineated, and current data linking ASD to gastrointestinal dysfunction have not been extensively reviewed. This Review outlines the established and emerging clinical and preclinical evidence that emphasizes the gut as a novel mechanistic and potential therapeutic target for individuals with ASD.
Collapse
Affiliation(s)
- Lin Y Hung
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA.
- Department of Cell Biology, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
- Department of Pediatrics, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
| |
Collapse
|
23
|
Lotakis DM, Rubalcava NS, Lopyan NM, Heider A, Rabah R, Elizabeth Speck K, Jarboe MD, Ehrlich PF, Ralls MW. Clinical relevance of inflammation on rectal biopsy for Hirschsprung disease: An outcomes analysis. J Pediatr Gastroenterol Nutr 2024; 78:36-42. [PMID: 38291694 DOI: 10.1002/jpn3.12047] [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: 04/04/2023] [Revised: 08/15/2023] [Accepted: 09/15/2023] [Indexed: 02/01/2024]
Abstract
OBJECTIVES Inflammation on diagnostic rectal biopsy for children with suspected Hirschsprung disease (HSCR) is reported on pathology, and its significance is unknown. We describe the management and outcomes of a cohort with inflammation on rectal biopsy compared to those without. Specifically, to address the hypothesis that inflammation on diagnostic biopsy is associated with increased complication rates irrespective of intervention type and timing. METHODS A single institution retrospective review of children with HSCR who underwent biopsy and endorectal pull-through (ERPT) from 2010 to 2020 was performed. The primary outcome was overall complications at 30-days following ERPT. Secondary outcomes included timing and type of operative intervention as well as postoperative enterocolitis diagnosed within 6-months of ERPT. RESULTS Forty-nine children were identified; inflammation was present on diagnostic biopsy for 17 children. Those with inflammation were more likely to have clinical evidence of enterocolitis at the time of biopsy (p = 0.001) and were more likely to undergo leveling colostomy before ERPT (p = 0.01). Children with inflammation had a higher anastomotic leak rate (p = 0.04). Subgroup analysis of patients with inflammation undergoing primary ERPT versus leveling colostomy demonstrated no significant difference in outcomes following definitive ERPT. CONCLUSIONS Our study suggests inflammation on diagnostic rectal biopsy for HSCR is associated with increased anastomotic leak rates. While additional prospective studies are indicated, attention to methods of mitigating inflammation and confirming its resolution before definitive pull-through may be of benefit for improving clinical outcomes in patients found with inflammation on diagnostic rectal biopsy.
Collapse
Affiliation(s)
- Dimitra M Lotakis
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Nathan S Rubalcava
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
- Department of Surgery, Creighton University Arizona Health Education Alliance, Phoenix, Arizona, USA
| | - Natalie M Lopyan
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Amer Heider
- Division of Pediatric and Perinatal Pathology, Department of Pathology, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Raja Rabah
- Division of Pediatric and Perinatal Pathology, Department of Pathology, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - K Elizabeth Speck
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Marcus D Jarboe
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Peter F Ehrlich
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| | - Matthew W Ralls
- Division of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Bonaz B. The gut-brain axis in Parkinson's disease. Rev Neurol (Paris) 2024; 180:65-78. [PMID: 38129277 DOI: 10.1016/j.neurol.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023]
Abstract
There is a bi-directional communication between the gut, including the microbiota, and the brain through the autonomic nervous system. Accumulating evidence has suggested a bidirectional link between gastrointestinal inflammation and neurodegeneration, in accordance with the concept of the gut-rain axis. An abnormal microbiota-gut-brain interaction contributes to the pathogeny of Parkinson's disease. This supports the hypothesis that Parkinson's disease originates in the gut to spread to the central nervous system, in particular through the vagus nerve. Targeting the gut-to-brain axis with vagus nerve stimulation, fecal microbiota transplantation, gut-selective antibiotics, as well as drugs targeting the leaky gut might be of interest in the management of Parkinson's disease.
Collapse
Affiliation(s)
- B Bonaz
- Service d'hépato-gastroentérologie, Grenoble institut neurosciences, université Grenoble-Alpes, Grenoble, France.
| |
Collapse
|
25
|
Wang X, Ding C, Li HB. The crosstalk between enteric nervous system and immune system in intestinal development, homeostasis and diseases. SCIENCE CHINA. LIFE SCIENCES 2024; 67:41-50. [PMID: 37672184 DOI: 10.1007/s11427-023-2376-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/19/2023] [Indexed: 09/07/2023]
Abstract
The gut is the largest digestive and absorptive organ, which is essential for induction of mucosal and systemic immune responses, and maintenance of metabolic-immune homeostasis. The intestinal components contain the epithelium, stromal cells, immune cells, and enteric nervous system (ENS), as well as the outers, such as gut microbiota, metabolites, and nutrients. The dyshomeostasis of intestinal microenvironment induces abnormal intestinal development and functions, even colon diseases including dysplasia, inflammation and tumor. Several recent studies have identified that ENS plays a crucial role in maintaining the immune homeostasis of gastrointestinal (GI) microenvironment. The crosstalk between ENS and immune cells, mainly macrophages, T cells, and innate lymphoid cells (ILCs), has been found to exert important regulatory roles in intestinal tissue programming, homeostasis, function, and inflammation. In this review, we mainly summarize the critical roles of the interactions between ENS and immune cells in intestinal homeostasis during intestinal development and diseases progression, to provide theoretical bases and ideas for the exploration of immunotherapy for gastrointestinal diseases with the ENS as potential novel targets.
Collapse
Affiliation(s)
- Xindi Wang
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenbo Ding
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hua-Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
26
|
Stolzer I, Scherer E, Süß P, Rothhammer V, Winner B, Neurath MF, Günther C. Impact of Microbiome-Brain Communication on Neuroinflammation and Neurodegeneration. Int J Mol Sci 2023; 24:14925. [PMID: 37834373 PMCID: PMC10573483 DOI: 10.3390/ijms241914925] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 09/27/2023] [Accepted: 10/01/2023] [Indexed: 10/15/2023] Open
Abstract
The gut microbiome plays a pivotal role in maintaining human health, with numerous studies demonstrating that alterations in microbial compositions can significantly affect the development and progression of various immune-mediated diseases affecting both the digestive tract and the central nervous system (CNS). This complex interplay between the microbiota, the gut, and the CNS is referred to as the gut-brain axis. The role of the gut microbiota in the pathogenesis of neurodegenerative diseases has gained increasing attention in recent years, and evidence suggests that gut dysbiosis may contribute to disease development and progression. Clinical studies have shown alterations in the composition of the gut microbiota in multiple sclerosis patients, with a decrease in beneficial bacteria and an increase in pro-inflammatory bacteria. Furthermore, changes within the microbial community have been linked to the pathogenesis of Parkinson's disease and Alzheimer's disease. Microbiota-gut-brain communication can impact neurodegenerative diseases through various mechanisms, including the regulation of immune function, the production of microbial metabolites, as well as modulation of host-derived soluble factors. This review describes the current literature on the gut-brain axis and highlights novel communication systems that allow cross-talk between the gut microbiota and the host that might influence the pathogenesis of neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Iris Stolzer
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Eveline Scherer
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Patrick Süß
- Department of Molecular Neurology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Veit Rothhammer
- Department of Neurology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| |
Collapse
|
27
|
Murray N, Al Khalaf S, Bastiaanssen TFS, Kaulmann D, Lonergan E, Cryan JF, Clarke G, Khashan AS, O’Connor K. Compositional and Functional Alterations in Intestinal Microbiota in Patients with Psychosis or Schizophrenia: A Systematic Review and Meta-analysis. Schizophr Bull 2023; 49:1239-1255. [PMID: 37210594 PMCID: PMC10483467 DOI: 10.1093/schbul/sbad049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
BACKGROUND AND HYPOTHESIS Intestinal microbiota is intrinsically linked to human health. Evidence suggests that the composition and function of the microbiome differs in those with schizophrenia compared with controls. It is not clear how these alterations functionally impact people with schizophrenia. We performed a systematic review and meta-analysis to combine and evaluate data on compositional and functional alterations in microbiota in patients with psychosis or schizophrenia. STUDY DESIGN Original studies involving humans and animals were included. The electronic databases PsycINFO, EMBASE, Web of Science, PubMed/MEDLINE, and Cochrane were systematically searched and quantitative analysis performed. STUDY RESULTS Sixteen original studies met inclusion criteria (1376 participants: 748 cases and 628 controls). Ten were included in the meta-analysis. Although observed species and Chao 1 show a decrease in diversity in people with schizophrenia compared with controls (SMD = -0.14 and -0.66 respectively), that did not reach statistical significance. We did not find evidence for variations in richness or evenness of microbiota between patients and controls overall. Differences in beta diversity and consistent patterns in microbial taxa were noted across studies. We found increases in Bifidobacterium, Lactobacillus, and Megasphaera in schizophrenia groups. Variations in brain structure, metabolic pathways, and symptom severity may be associated with compositional alterations in the microbiome. The heterogeneous design of studies complicates a similar evaluation of functional readouts. CONCLUSIONS The microbiome may play a role in the etiology and symptomatology of schizophrenia. Understanding how the implications of alterations in microbial genes for symptomatic expression and clinical outcomes may contribute to the development of microbiome targeted interventions for psychosis.
Collapse
Affiliation(s)
- Nuala Murray
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Sukainah Al Khalaf
- School of Public Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - David Kaulmann
- School of Public Health, University College Cork, Cork, Ireland
| | - Edgar Lonergan
- RISE, Early Intervention in Psychosis Service, South Lee Mental Health Services, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ali S Khashan
- School of Public Health, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| | - Karen O’Connor
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
- RISE, Early Intervention in Psychosis Service, South Lee Mental Health Services, Cork, Ireland
| |
Collapse
|
28
|
Huang Y, Guo Y, Li X, Xiao Y, Wang Z, Song L, Ren Z. Effects of Lactiplantibacillus plantarum GUANKE on Diphenoxylate-Induced Slow Transit Constipation and Gut Microbiota in Mice. Nutrients 2023; 15:3741. [PMID: 37686774 PMCID: PMC10490327 DOI: 10.3390/nu15173741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Slow transit constipation (STC) is a prevalent gastrointestinal condition with slow transit, and some probiotics can effectively relieve constipation, but the exact mechanisms have not been fully understood. In this study, we evaluate the impact of Lactiplantibacillus plantarum GUANKE (GUANKE) on diphenoxylate-induced slow transit constipation and speculate on the underlying mechanisms in a mouse model. Administration of L. plantarum GUANKE alleviated constipation indexes, including defecation time, fecal output and water content, and gastrointestinal transit ratio. In addition, GUANKE restored the protein expression of constipation-related intestinal factors (aquaporins (AQPs) and interstitial Cajal cells (ICCs)) in colon tissues measured using immunofluorescence staining; regulated the neurotransmitters and hormones, such as increased levels of 5-hydroxytryptamine, substance P, and motilin; and decreased levels of vasoactive intestinal peptide and nitric oxide in serum, as measured by an ELISA. 16S rRNA and correlation analysis of feces indicated that GUANKE administration effectively reduced constipation-induced Prevotella enrichment and suggested a potential contribution of Prevotella to diphenoxylate-induced STC in mice. GUANKE had no effect on short-chain fatty acids (SCFAs) in cecum content. This study revealed that GUANKE may alleviate constipation in mice through regulating intestinal neurotransmitter and hormone release and altering specific bacterial taxa, rather than by affecting SCFAs and the diversity of microbiota in the gut. Further research is needed to confirm if the findings observed in this study will be consistent in other animal studies or clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Liqiong Song
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases Chinese Center for Disease Control and Prevention, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (Y.H.); (Y.G.); (X.L.); (Y.X.); (Z.W.)
| | - Zhihong Ren
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases Chinese Center for Disease Control and Prevention, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China; (Y.H.); (Y.G.); (X.L.); (Y.X.); (Z.W.)
| |
Collapse
|
29
|
Wang X, Tang R, Wei Z, Zhan Y, Lu J, Li Z. The enteric nervous system deficits in autism spectrum disorder. Front Neurosci 2023; 17:1101071. [PMID: 37694110 PMCID: PMC10484716 DOI: 10.3389/fnins.2023.1101071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Gastrointestinal (GI) disorders are common comorbidities in individuals with autism spectrum disorder (ASD), and abnormalities in these issues have been found to be closely related to the severity of core behavioral deficits in autism. The enteric nervous system (ENS) plays a crucial role in regulating various aspects of gut functions, including gastrointestinal motility. Dysfunctional wiring in the ENS not only results in various gastrointestinal issues, but also correlates with an increasing number of central nervous system (CNS) disorders, such as ASD. However, it remains unclear whether the gastrointestinal dysfunctions are a consequence of ASD or if they directly contribute to its pathogenesis. This review focuses on the deficits in the ENS associated with ASD, and highlights several high-risk genes for ASD, which are expressed widely in the gut and implicated in gastrointestinal dysfunction among both animal models and human patients with ASD. Furthermore, we provide a brief overview of environmental factors associated with gastrointestinal tract in individuals with autism. This could offer fresh perspectives on our understanding of ASD.
Collapse
Affiliation(s)
- Xinnian Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Life Science, USTC Life Sciences and Medicine, Hefei, China
| | - Ruijin Tang
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Wei
- Department of Child Psychiatry and Rehabilitation, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Yang Zhan
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianping Lu
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
| | - Zhiling Li
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
30
|
Lefèvre MA, Soret R, Pilon N. Harnessing the Power of Enteric Glial Cells' Plasticity and Multipotency for Advancing Regenerative Medicine. Int J Mol Sci 2023; 24:12475. [PMID: 37569849 PMCID: PMC10419543 DOI: 10.3390/ijms241512475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
The enteric nervous system (ENS), known as the intrinsic nervous system of the gastrointestinal tract, is composed of a diverse array of neuronal and glial cell subtypes. Fascinating questions surrounding the generation of cellular diversity in the ENS have captivated ENS biologists for a considerable time, particularly with recent advancements in cell type-specific transcriptomics at both population and single-cell levels. However, the current focus of research in this field is predominantly restricted to the study of enteric neuron subtypes, while the investigation of enteric glia subtypes significantly lags behind. Despite this, enteric glial cells (EGCs) are increasingly recognized as equally important regulators of numerous bowel functions. Moreover, a subset of postnatal EGCs exhibits remarkable plasticity and multipotency, distinguishing them as critical entities in the context of advancing regenerative medicine. In this review, we aim to provide an updated overview of the current knowledge on this subject, while also identifying key questions that necessitate future exploration.
Collapse
Affiliation(s)
- Marie A. Lefèvre
- Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montreal, QC H3C 3P8, Canada;
- Centre D’excellence en Recherche Sur Les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Rodolphe Soret
- Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montreal, QC H3C 3P8, Canada;
- Centre D’excellence en Recherche Sur Les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
| | - Nicolas Pilon
- Département des Sciences Biologiques, Université du Québec à Montréal (UQAM), Montreal, QC H3C 3P8, Canada;
- Centre D’excellence en Recherche Sur Les Maladies Orphelines—Fondation Courtois (CERMO-FC), Université du Québec à Montréal, Montreal, QC H2X 3Y7, Canada
- Département de Pédiatrie, Université de Montréal, Montreal, QC H3T 1C5, Canada
| |
Collapse
|
31
|
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
|
32
|
De Sales-Millán A, Aguirre-Garrido JF, González-Cervantes RM, Velázquez-Aragón JA. Microbiome-Gut-Mucosal-Immune-Brain Axis and Autism Spectrum Disorder (ASD): A Novel Proposal of the Role of the Gut Microbiome in ASD Aetiology. Behav Sci (Basel) 2023; 13:548. [PMID: 37503995 PMCID: PMC10376175 DOI: 10.3390/bs13070548] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder characterised by deficits in social interaction and communication, as well as restricted and stereotyped interests. Due of the high prevalence of gastrointestinal disorders in individuals with ASD, researchers have investigated the gut microbiota as a potential contributor to its aetiology. The relationship between the microbiome, gut, and brain (microbiome-gut-brain axis) has been acknowledged as a key factor in modulating brain function and social behaviour, but its connection to the aetiology of ASD is not well understood. Recently, there has been increasing attention on the relationship between the immune system, gastrointestinal disorders and neurological issues in ASD, particularly in relation to the loss of specific species or a decrease in microbial diversity. It focuses on how gut microbiota dysbiosis can affect gut permeability, immune function and microbiota metabolites in ASD. However, a very complete study suggests that dysbiosis is a consequence of the disease and that it has practically no effect on autistic manifestations. This is a review of the relationship between the immune system, microbial diversity and the microbiome-gut-brain axis in the development of autistic symptoms severity and a proposal of a novel role of gut microbiome in ASD, where dysbiosis is a consequence of ASD-related behaviour and where dysbiosis in turn accentuates the autistic manifestations of the patients via the microbiome-gut-brain axis in a feedback circuit.
Collapse
Affiliation(s)
- Amapola De Sales-Millán
- División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Lerma, Lerma 52006, Estado de Mexico, Mexico
| | - José Félix Aguirre-Garrido
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma 52006, Estado de Mexico, Mexico
| | - Rina María González-Cervantes
- Departamento de Ciencias Ambientales, Universidad Autónoma Metropolitana-Lerma, Lerma 52006, Estado de Mexico, Mexico
| | | |
Collapse
|
33
|
Caetano MAF, Magalhães HIR, Duarte JRL, Conceição LB, Castelucci P. Butyrate Protects Myenteric Neurons Loss in Mice Following Experimental Ulcerative Colitis. Cells 2023; 12:1672. [PMID: 37443707 PMCID: PMC10340616 DOI: 10.3390/cells12131672] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/08/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
The enteric nervous system is affected by inflammatory bowel diseases (IBD). Gut microbiota ferments dietary fibers and produces short-chain fatty acids, such as Butyrate, which bind to G protein-coupled receptors, such as GPR41, and contribute to maintaining intestinal health. This work aimed to study the GPR41 in myenteric neurons and analyze the effect of Butyrate in mice submitted to experimental ulcerative colitis. The 2, 4, 6 trinitrobenzene sulfonic acid (TNBS) was injected intrarectally in C57BL/6 mice (Colitis). Sham group received ethanol (vehicle). One group was treated with 100 mg/kg of Sodium Butyrate (Butyrate), and the other groups received saline. Animals were euthanized 7 days after colitis induction. Analyzes demonstrated colocalization of GPR41 with neurons immunoreactive (-ir) to nNOS and ChAT-ir and absence of colocalization of the GPR41 with GFAP-ir glia. Quantitative results demonstrated losses of nNOS-ir, ChAT-ir, and GPR41-ir neurons in the Colitis group and Butyrate treatment attenuated neuronal loss. The number of GFAP-ir glia increased in the Colitis group, whereas Butyrate reduced the number of these cells. In addition, morphological alterations observed in the Colitis group were attenuated in the Butyrate group. The presence of GPR41 in myenteric neurons was identified, and the treatment with Butyrate attenuated the damage caused by experimental ulcerative colitis.
Collapse
Affiliation(s)
- Marcos A. F. Caetano
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.A.F.C.); (J.R.L.D.); (L.B.C.)
| | - Henrique I. R. Magalhães
- Department of Surgery, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 05508-270, Brazil;
| | - Jheniffer R. L. Duarte
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.A.F.C.); (J.R.L.D.); (L.B.C.)
| | - Laura B. Conceição
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.A.F.C.); (J.R.L.D.); (L.B.C.)
| | - Patricia Castelucci
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (M.A.F.C.); (J.R.L.D.); (L.B.C.)
| |
Collapse
|
34
|
Stein RA, Riber L. Epigenetic effects of short-chain fatty acids from the large intestine on host cells. MICROLIFE 2023; 4:uqad032. [PMID: 37441522 PMCID: PMC10335734 DOI: 10.1093/femsml/uqad032] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/04/2023] [Accepted: 06/14/2023] [Indexed: 07/15/2023]
Abstract
Adult humans harbor at least as many microbial cells as eukaryotic ones. The largest compartment of this diverse microbial population, the gut microbiota, encompasses the collection of bacteria, archaea, viruses, and eukaryotic organisms that populate the gastrointestinal tract, and represents a complex and dynamic ecosystem that has been increasingly implicated in health and disease. The gut microbiota carries ∼100-to-150-times more genes than the human genome and is intimately involved in development, homeostasis, and disease. Of the several microbial metabolites that have been studied, short-chain fatty acids emerge as a group of molecules that shape gene expression in several types of eukaryotic cells by multiple mechanisms, which include DNA methylation changes, histone post-translational modifications, and microRNA-mediated gene silencing. Butyric acid, one of the most extensively studied short-chain fatty acids, reaches higher concentrations in the colonic lumen, where it provides a source of energy for healthy colonocytes, and its concentrations decrease towards the bottom of the colonic crypts, where stem cells reside. The lower butyric acid concentration in the colonic crypts allows undifferentiated cells, such as stem cells, to progress through the cell cycle, pointing towards the importance of the crypts in providing them with a protective niche. In cancerous colonocytes, which metabolize relatively little butyric acid and mostly rely on glycolysis, butyric acid preferentially acts as a histone deacetylase inhibitor, leading to decreased cell proliferation and increased apoptosis. A better understanding of the interface between the gut microbiota metabolites and epigenetic changes in eukaryotic cells promises to unravel in more detail processes that occur physiologically and as part of disease, help develop novel biomarkers, and identify new therapeutic modalities.
Collapse
Affiliation(s)
- Richard A Stein
- Corresponding author. Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, 6 MetroTech Center, Brooklyn, NY 11201, USA. Tel: +1-917-684-9438; E-mail: ;
| | - Leise Riber
- Department of Plant & Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg, Denmark
| |
Collapse
|
35
|
Zhao L, Xiao J, Li S, Guo Y, Fu R, Hua S, Du Y, Xu S. The interaction between intestinal microenvironment and stroke. CNS Neurosci Ther 2023; 29 Suppl 1:185-199. [PMID: 37309254 PMCID: PMC10314114 DOI: 10.1111/cns.14275] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Stroke is not only a major cause of disability but also the third leading cause of death, following heart disease and cancer. It has been established that stroke causes permanent disability in 80% of survivors. However, current treatment options for this patient population are limited. Inflammation and immune response are major features that are well-recognized to occur after a stroke. The gastrointestinal tract hosts complex microbial communities, the largest pool of immune cells, and forms a bidirectional regulation brain-gut axis with the brain. Recent experimental and clinical studies have highlighted the importance of the relationship between the intestinal microenvironment and stroke. Over the years, the influence of the intestine on stroke has emerged as an important and dynamic research direction in biology and medicine. AIMS In this review, we describe the structure and function of the intestinal microenvironment and highlight its cross-talk relationship with stroke. In addition, we discuss potential strategies aiming to target the intestinal microenvironment during stroke treatment. CONCLUSION The structure and function of the intestinal environment can influence neurological function and cerebral ischemic outcome. Improving the intestinal microenvironment by targeting the gut microbiota may be a new direction in treating stroke.
Collapse
Affiliation(s)
- Linna Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Translational Research of TCM Prescription and SyndromeTianjinChina
| | - Jie Xiao
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Songlin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Yuying Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Translational Research of TCM Prescription and SyndromeTianjinChina
| | - Rong Fu
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Shengyu Hua
- Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Yuzheng Du
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
| | - Shixin Xu
- First Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Translational Research of TCM Prescription and SyndromeTianjinChina
| |
Collapse
|
36
|
Seguella L, Palenca I, Franzin SB, Zilli A, Esposito G. Mini-review: Interaction between intestinal microbes and enteric glia in health and disease. Neurosci Lett 2023; 806:137221. [PMID: 37031943 DOI: 10.1016/j.neulet.2023.137221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/11/2023]
Abstract
Enteric glia are a unique population of peripheral neuroglia associated with the enteric nervous system (ENS) throughout the digestive tract. The emerging data from the latest glial biology studies unveiled enteric glia as a heterogenic population with plastic and adaptative abilities that display phenotypic and functional changes upon distinct extrinsic cues. This aspect is essential in the dynamic signaling that enteric glia engage with neurons and other neighboring cells within the intestinal wall, such as epithelial, endocrine, and immune cells to maintain local homeostasis. Likewise, enteric glia sense signals from luminal microbes, although the extent of this active communication is still unclear. In this minireview, we discuss the recent findings that support glia-microbes crosstalk in the intestine in health and disease, pointing out the critical aspects that require further investigation.
Collapse
Affiliation(s)
- Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy.
| | - Irene Palenca
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy.
| | - Silvia Basili Franzin
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy.
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy.
| | - Giuseppe Esposito
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Italy.
| |
Collapse
|
37
|
Calabrò S, Kankowski S, Cescon M, Gambarotta G, Raimondo S, Haastert-Talini K, Ronchi G. Impact of Gut Microbiota on the Peripheral Nervous System in Physiological, Regenerative and Pathological Conditions. Int J Mol Sci 2023; 24:ijms24098061. [PMID: 37175764 PMCID: PMC10179357 DOI: 10.3390/ijms24098061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
It has been widely demonstrated that the gut microbiota is responsible for essential functions in human health and that its perturbation is implicated in the development and progression of a growing list of diseases. The number of studies evaluating how the gut microbiota interacts with and influences other organs and systems in the body and vice versa is constantly increasing and several 'gut-organ axes' have already been defined. Recently, the view on the link between the gut microbiota (GM) and the peripheral nervous system (PNS) has become broader by exceeding the fact that the PNS can serve as a systemic carrier of GM-derived metabolites and products to other organs. The PNS as the communication network between the central nervous system and the periphery of the body and internal organs can rather be affected itself by GM perturbation. In this review, we summarize the current knowledge about the impact of gut microbiota on the PNS, with regard to its somatic and autonomic divisions, in physiological, regenerative and pathological conditions.
Collapse
Affiliation(s)
- Sonia Calabrò
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Svenja Kankowski
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Kirsten Haastert-Talini
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover (ZSN), Buenteweg 2, 30559 Hannover, Germany
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| |
Collapse
|
38
|
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
|
39
|
Costa CJ, Cohen MW, Goldberg DC, Mellado W, Willis DE. Nicotinamide Riboside Improves Enteric Neuropathy in Streptozocin-Induced Diabetic Rats Through Myenteric Plexus Neuroprotection. Dig Dis Sci 2023:10.1007/s10620-023-07913-5. [PMID: 36920665 DOI: 10.1007/s10620-023-07913-5] [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: 05/31/2022] [Accepted: 03/03/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Diabetes Mellitus causes a systemic oxidative stress due in part to the hyperglycemia and the reactive oxygen species generated. Up to 75% of diabetic patients present with an autonomic neuropathy affecting the Enteric Nervous System. Deficits in the human population are chronic dysmotilities with either increased (i.e., constipation) or decreased (i.e., diarrhea) total gastrointestinal transit times. These are recapitulated in the streptozocin-induced diabetic rat, which is a model of Type I Diabetes Mellitus. AIMS Examine the effects that a precursor of nicotinamide adenosine dinucleotide (NAD), nicotinamide riboside (NR), had on the development of dysmotility in induced diabetic rats and if fecal microbiota transplant (FMT) could produce the same results. MATERIALS AND METHODS Utilizing a 6-week treatment paradigm, NR was administered intraperitoneally every 48 h. Total gastrointestinal transit time was assessed weekly utilizing the carmine red method. Three weeks following hyperglycemic induction, FMT was performed between NR-treated animals and untreated animals. SIGNIFICANT RESULTS There is improvement in overall gastrointestinal transit time with the use of NR. 16S microbiome sequencing demonstrated decreased alpha and beta diversity in induced diabetic rats without change in animals receiving FMT. Improvements in myenteric plexus ganglia density in small and large intestines in diabetic animals treated with NR were seen. CONCLUSIONS NR treatment led to functional improvement in total gastrointestinal transit time in induced diabetic animals. This was associated with neuroprotection in the myenteric plexuses of both small and large intestines of induced diabetic rats. This represents an important first step in showing NR's benefit as a treatment for diabetic enteric neuropathy. Streptozocin-induced diabetic rats have improved transit times and increased myenteric plexus ganglia density when treated with intraperitoneal nicotinamide riboside.
Collapse
Affiliation(s)
- Christopher J Costa
- Quinnipiac University Frank H Netter MD School of Medicine, North Haven, CT, USA. .,Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA. .,Graduate Medical Education, Internal Medicine Residency, UConn Health, 263 Farmington Ave, Farmington, CT, 06030-1235, USA.
| | - Melanie W Cohen
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA
| | - David C Goldberg
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA.,Children's Hospital of Philadelphia, 3501 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Wilfredo Mellado
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA
| | - Dianna E Willis
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY, 10605, USA.,Weill Cornell Medicine Feil Family Brain and Mind Research Institute, New York, NY, USA
| |
Collapse
|
40
|
Mo K, Yu W, Li J, Zhang Y, Xu Y, Huang X, Ni H. Dietary supplementation with a microencapsulated complex of thymol, carvacrol, and cinnamaldehyde improves intestinal barrier function in weaning piglets. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:1994-2003. [PMID: 36347590 DOI: 10.1002/jsfa.12322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/19/2022] [Accepted: 11/09/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The authors previously prepared a microencapsulated complex of thymol, carvacrol, and cinnamaldehyde (MEEO). This study aimed to evaluate the effect of MEEO on the intestinal mucosal barrier and homeostasis in weaning piglets. A comparison of the effect of MEEO versus chlortetracycline (CTC) was performed in this study. RESULTS Piglets were divided into three groups - control (Con), MEEO, and CTC groups - and raised for 28 days. The results showed that MEEO significantly elevated the ratio of the villus height and the crypt depth in the jejunum and decreased the crypt depth in the ileum compared with the other groups (P < 0.05); it also upregulated the messenger ribonucleic acid (mRNA) expression of tight junction protein in the small intestine. Compared with the Con group, MEEO increased the concentration of secretory immunoglobulin A (sIgA), cathelicidin antimicrobial peptides (CAMP), and interleukin 10 (IL-10), while decreasing the interleukin 1 beta (IL-1β) concentration in both jejunal and ileal mucosa (P < 0.05). The mRNA expression of jejunal mucosal MUC1 and ileal mucosal MUC2 was increased in the MEEO group compared with the other groups (P < 0.05). Intestinal microbial analysis showed that dietary treatment had little impact on the ileal microbial structure. A significant rise in the genus Lactobacillus was, however, found in the MEEO group. There is a positive correlation between the Lactobacillus and sIgA, and between the Lactobacillus and CAMP, indicating that an improvement in the mucosal barrier function by the addition of MEEO may be associated with the proliferation of Lactobacillus. CONCLUSION Dietary supplementation with MEEO improves intestinal barrier function in weaning piglets, the effect of which was superior to CTC. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Kaibin Mo
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wengang Yu
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jing Li
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yunxiao Zhang
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Xu
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xianhui Huang
- Guangdong Key Laboratory for Veterinary Drug Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hengjia Ni
- National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Province Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
41
|
Immunity orchestrates a bridge in gut-brain axis of neurodegenerative diseases. Ageing Res Rev 2023; 85:101857. [PMID: 36669690 DOI: 10.1016/j.arr.2023.101857] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Neurodegenerative diseases, in particular for Alzheimer's disease (AD), Parkinson's disease (PD) and Multiple sclerosis (MS), are a category of diseases with progressive loss of neuronal structure or function (encompassing neuronal death) leading to neuronal dysfunction, whereas the underlying pathogenesis remains to be clarified. As the microbiological ecosystem of the intestinal microbiome serves as the second genome of the human body, it is strongly implicated as an essential element in the initiation and/or progression of neurodegenerative diseases. Nevertheless, the precise underlying principles of how the intestinal microflora impact on neurodegenerative diseases via gut-brain axis by modulating the immune function are still poorly characterized. Consequently, an overview of initiating the development of neurodegenerative diseases and the contribution of intestinal microflora on immune function is discussed in this review.
Collapse
|
42
|
The Potential Role of Microorganisms on Enteric Nervous System Development and Disease. Biomolecules 2023; 13:biom13030447. [PMID: 36979382 PMCID: PMC10046024 DOI: 10.3390/biom13030447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/14/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The enteric nervous system (ENS), the inherent nervous system of the gastrointestinal (GI) tract is a vast nervous system that controls key GI functions, including motility. It functions at a critical interface between the gut luminal contents, including the diverse population of microorganisms deemed the microbiota, as well as the autonomic and central nervous systems. Critical development of this axis of interaction, a key determinant of human health and disease, appears to occur most significantly during early life and childhood, from the pre-natal through to the post-natal period. These factors that enable the ENS to function as a master regulator also make it vulnerable to damage and, in turn, a number of GI motility disorders. Increasing attention is now being paid to the potential of disruption of the microbiota and pathogenic microorganisms in the potential aetiopathogeneis of GI motility disorders in children. This article explores the evidence regarding the relationship between the development and integrity of the ENS and the potential for such factors, notably dysbiosis and pathogenic bacteria, viruses and parasites, to impact upon them in early life.
Collapse
|
43
|
Lotakis DM, Dheer R, Dame MK, Cuttitta AJ, Tigani DJ, Spence JR, Young VB, Ralls MW. A Pilot Study: Transcriptional Profiling, Functional Analysis, and Organoid Modeling of Intestinal Mucosa in Hirschsprung Disease. J Pediatr Surg 2023; 58:1164-1169. [PMID: 36922279 DOI: 10.1016/j.jpedsurg.2023.02.020] [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: 01/22/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Hirschsprung disease (HSCR) is a congenital colonic aganglionosis. Many HSCR patients develop enterocolitis despite surgical resection. The pathophysiology of this inflammatory process is poorly understood. We compared transcriptional profiles and function of ganglionic and aganglionic tissue in HSCR patients. METHODS RNA sequencing was performed on mucosal tissues from HSCR patients (n = 6) and controls (n = 3). Function of matched ganglionic and aganglionic regions were investigated utilizing organoids generated from these tissues. RESULTS Transcriptional differences observed in ganglionic and aganglionic regions of HSCR patients included upregulation of genes involving inflammation, cell differentiation and proliferation as well as decreased expression of genes encoding mucins compared to controls. Organoids derived from ganglionic and aganglionic regions of HSCR patients were similar in epithelial cell differentiation, epithelial barrier formation and response to stimulation with bacterial metabolites and pro-inflammatory cytokines. CONCLUSIONS Despite normal ganglionic structure, the section of colon adjacent to the aganglionic region in HSCR patients has perturbed gene expression which resembles the aganglionic segment. Transcriptional and functional changes in colonic epithelium are persevered in the ganglionic colon used for pull-through surgery. This may explain persistence of enterocolitis despite surgical excision of aganglionic colon and subsequent endorectal pull-through performed with ganglionic colon during correction of HSCR. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Dimitra M Lotakis
- University of Michigan, Department of Surgery, Division of Pediatric Surgery, 1540 East Hospital Drive, Ann Arbor, MI, 48109-4211, USA.
| | - Rishu Dheer
- University of Michigan, Department of Internal Medicine, Division of Infectious Disease, Ann Arbor, MI, 48109, USA
| | - Michael K Dame
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, Ann Arbor, MI, 48109, USA
| | - Ashley J Cuttitta
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, Ann Arbor, MI, 48109, USA
| | - Dominic J Tigani
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, Ann Arbor, MI, 48109, USA
| | - Jason R Spence
- University of Michigan, Department of Internal Medicine, Division of Gastroenterology, Ann Arbor, MI, 48109, USA; University of Michigan Medical School, Department of Cell and Developmental Biology, Ann Arbor, MI, 48109, USA; University of Michigan Medical School, Department of Biomedical Engineering Medicine, Ann Arbor, MI, 48109, USA
| | - Vincent B Young
- University of Michigan, Department of Internal Medicine, Division of Infectious Disease, Ann Arbor, MI, 48109, USA; University of Michigan, Department of Microbiology & Immunology, Ann Arbor, MI, 48109, USA
| | - Matthew W Ralls
- University of Michigan, Department of Surgery, Division of Pediatric Surgery, 1540 East Hospital Drive, Ann Arbor, MI, 48109-4211, USA
| |
Collapse
|
44
|
Kasarello K, Cudnoch-Jedrzejewska A, Czarzasta K. Communication of gut microbiota and brain via immune and neuroendocrine signaling. Front Microbiol 2023; 14:1118529. [PMID: 36760508 PMCID: PMC9907780 DOI: 10.3389/fmicb.2023.1118529] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
The gastrointestinal tract of the human is inhabited by about 5 × 1013 bacteria (of about 1,000 species) as well as archaea, fungi, and viruses. Gut microbiota is known to influence the host organism, but the host may also affect the functioning of the microbiota. This bidirectional cooperation occurs in three main inter-organ signaling: immune, neural, and endocrine. Immune communication relies mostly on the cytokines released by the immune cells into circulation. Also, pathogen-associated or damage-associated molecular patterns (PAMPs or DAMPs) may enter circulation and affect the functioning of the internal organs and gut microbiota. Neural communication relies mostly on the direct anatomical connections made by the vagus nerve, or indirect connections via the enteric nervous system. The third pathway, endocrine communication, is the broadest one and includes the hypothalamic-pituitary-adrenal axis. This review focuses on presenting the latest data on the role of the gut microbiota in inter-organ communication with particular emphasis on the role of neurotransmitters (catecholamines, serotonin, gamma-aminobutyric acid), intestinal peptides (cholecystokinin, peptide YY, and glucagon-like peptide 1), and bacterial metabolites (short-chain fatty acids).
Collapse
|
45
|
Baghdadi MB, Kim TH. The multiple roles of enteric glial cells in intestinal homeostasis and regeneration. Semin Cell Dev Biol 2023:S1084-9521(23)00005-8. [PMID: 36658046 DOI: 10.1016/j.semcdb.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 12/16/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
The gastrointestinal tract is innervated by the enteric nervous system (ENS), a complex network of neurons and glial cells, also called the "second brain". Enteric glial cells, one of the major cell types in the ENS, are located throughout the entire gut wall. Accumulating evidence has demonstrated their critical requirement for gut physiology. Notably, recent studies have shown that enteric glial cells control new aspects of gut function such as regulation of intestinal stem cell behavior and immunity. In addition, the emergence of single-cell genomics technologies has revealed enteric glial cell heterogeneity and plasticity. In this review, we discuss established and emerging concepts regarding the roles of mammalian enteric glial cells and their heterogeneity in gut development, homeostasis, and regeneration.
Collapse
Affiliation(s)
- Meryem B Baghdadi
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Tae-Hee Kim
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
46
|
Ruigrok RAAA, Weersma RK, Vich Vila A. The emerging role of the small intestinal microbiota in human health and disease. Gut Microbes 2023; 15:2201155. [PMID: 37074215 PMCID: PMC10120449 DOI: 10.1080/19490976.2023.2201155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/03/2023] [Indexed: 04/20/2023] Open
Abstract
The human gut microbiota continues to demonstrate its importance in human health and disease, largely owing to the countless number of studies investigating the fecal microbiota. Underrepresented in these studies, however, is the role played by microbial communities found in the small intestine, which, given the essential function of the small intestine in nutrient absorption, host metabolism, and immunity, is likely highly relevant. This review provides an overview of the methods used to study the microbiota composition and dynamics along different sections of the small intestine. Furthermore, it explores the role of the microbiota in facilitating the small intestine in its physiological functions and discusses how disruption of the microbial equilibrium can influence disease development. The evidence suggests that the small intestinal microbiota is an important regulator of human health and its characterization has the potential to greatly advance gut microbiome research and the development of novel disease diagnostics and therapeutics.
Collapse
Affiliation(s)
- Renate A. A. A. Ruigrok
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
47
|
Electroacupuncture Modulates 5-HT 4R-Mediated cAMP/PKA Signaling to Improve Intestinal Motility Disorders in a Thy1- αSyn Parkinson's Mouse Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8659462. [PMID: 36337584 PMCID: PMC9635967 DOI: 10.1155/2022/8659462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/02/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
Constipation is one of the most common nonmotor symptoms in patients with Parkinson's disease (PD) and often occurs before motor symptoms. Electroacupuncture effectively improves the symptoms of constipation in patients with PD. In the present study, we used thymus cell antigen 1-α-synuclein (Thy1-αSyn) transgenic mice as a model of intestinal motility disorders in PD to determine the therapeutic effect of electroacupuncture and the underlying mechanisms. Electroacupuncture significantly improved fecal excretion and accelerated the rate of small-intestinal propulsion in Thy1-αSyn mice by upregulating the serotonin concentration and the expression of the serotonin 4 receptor. Consequently, the downstream cyclic AMP/protein kinase A (cAMP/PKA) pathway was affected, and to upregulate and downregulate, the expression of substance P was upregulated, and the expression of calcitonin gene-related peptide was downregulated. In summary, electroacupuncture improved intestinal motility in PD mice by affecting serotonin levels, serotonin 4 receptor expression, and the cAMP/PKA pathway, providing a potentially effective and promising complementary and alternative therapy for relieving constipation symptoms in patients with PD.
Collapse
|
48
|
Caetano MAF, Castelucci P. Role of short chain fatty acids in gut health and possible therapeutic approaches in inflammatory bowel diseases. World J Clin Cases 2022; 10:9985-10003. [PMID: 36246826 PMCID: PMC9561599 DOI: 10.12998/wjcc.v10.i28.9985] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/02/2022] [Accepted: 08/25/2022] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by inflammation in the gastrointestinal tract and include Ulcerative Colitis and Crohn’s Disease. These diseases are costly to health services, substantially reduce patients’ quality of life, and can lead to complications such as cancer and even death. Symptoms include abdominal pain, stool bleeding, diarrhea, and weight loss. The treatment of these diseases is symptomatic, seeking disease remission. The intestine is colonized by several microorganisms, such as fungi, viruses, and bacteria, which constitute the intestinal microbiota (IM). IM bacteria promotes dietary fibers fermentation and produces short-chain fatty acids (SCFAs) that exert several beneficial effects on intestinal health. SCFAs can bind to G protein-coupled receptors, such as GPR41 and GPR43, promoting improvements in the intestinal barrier, anti-inflammatory, and antioxidant effects. Thus, SCFAs could be a therapeutic tool for IBDs. However, the mechanisms involved in these beneficial effects of SCFAs remain poorly understood. Therefore, this paper aims to provide a review addressing the main aspects of IBDs, and a more detailed sight of SCFAs, focusing on the main effects on different aspects of the intestine with an emphasis on IBDs.
Collapse
Affiliation(s)
| | - Patricia Castelucci
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508900, SP, Brazil
| |
Collapse
|
49
|
Gut microbiota: a new avenue to reveal pathological mechanisms of constipation. Appl Microbiol Biotechnol 2022; 106:6899-6913. [PMID: 36190540 DOI: 10.1007/s00253-022-12197-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
Constipation is very pervasive all over the world. It is a common multifactorial gastrointestinal disease, and its etiology and pathomechanism are not completely clear. Now, increasing evidence shows that intestinal flora is closely related to constipation. Intestinal flora is the largest microbiota in the human body and has powerful metabolic functions. Intestinal flora can produce a variety of metabolites, such as bile acids, short-chain fatty acids, tryptophan metabolites, and methane, which have important effects on intestinal motility and secretion. The host can also monitor the intestinal flora and regulate gut dysbacteriosis in constipation. To explore the relationship between intestinal flora and host, the combination of multiomics technology has become the powerful and effective method. Furthermore, the homeostasis restoration of intestinal flora also provides a new strategy for the treatment of constipation. This review aims to explore the interaction between intestinal flora and host in constipation, which contributes to disclose the pathogenesis of constipation and the development of novel drugs for the treatment of constipation from the perspective of intestinal flora. KEY POINTS: • This review highlights the regulation of gut microbiota on the intestinal motility and secretion of host. • The current review gives an insight into the role of the host on the recognition and regulation of intestinal ecology under constipation. • The article also introduces some novel methods of current gut microbiota research and gut microbiota-based constipation therapies.
Collapse
|
50
|
Wu PN, Xiong S, Zhong P, Yang WQ, Chen M, Tang TC. Global trends in research on irritable bowel syndrome and the brain–gut axis: Bibliometrics and visualization analysis. Front Pharmacol 2022; 13:956204. [PMID: 36160395 PMCID: PMC9493189 DOI: 10.3389/fphar.2022.956204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a gastrointestinal disorder with no structural damage, and its pathogenesis remains unclear. Studies have shown that the brain–gut axis is closely related to the occurrence of IBS. However, studies of IBS related to the brain–gut axis have not been systematically analyzed by bibliometrics and visual analysis. This study is based on 631 publications in the Web of Science Core Collection (WoSCC) to analyze hot spots and trends in this field. The collaborations between different authors, institutions, countries, and keywords were bibliometrically analyzed by CiteSpace software. Meanwhile, VOSviewer analyzed the references. The results show that since 2012, the number of publications has been growing rapidly. According to the collaborative network analysis, the United States, the National University of Ireland, Cork, and J.F. Cryan are the countries, institutions, and authors contributing the most, respectively. Through keywords and literature analysis, mechanisms and therapy associated with IBS and the brain–gut axis have still been a research focus in recent years. Furthermore, the physiological and pathological mechanisms of the brain–gut axis influencing IBS (related to gastrointestinal dysfunction, vagus nerve, visceral pain, intestinal flora, serotonin, tryptophan metabolism, stress, brain-derived neurotrophic factor (BDNF), and malonyldialdehyde) are the future research trends, especially the mechanisms related to intestinal flora. This is the first bibliometric and visualization analysis of IBS and brain–gut axis-related literature to explore research hotspots and trends.
Collapse
Affiliation(s)
- Peng-Ning Wu
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Shuai Xiong
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Peng Zhong
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Wan-Qing Yang
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Min Chen
- Department of colorectal diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tai-Chun Tang
- Department of colorectal diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Tai-Chun Tang,
| |
Collapse
|