Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Intervalo de año de publicación
1.
J Autoimmun ; 148: 103292, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39067313

RESUMEN

Disruption of gut barrier function and intestinal immune cell homeostasis are increasingly considered critical players in pathogenesis of extra-intestinal inflammatory diseases, including multiple sclerosis (MS) and its prototypical animal model, the experimental autoimmune encephalomyelitis (EAE). Breakdown of epithelial barriers increases intestinal permeability and systemic dissemination of microbiota-derived molecules. However, whether the gut-vascular barrier (GVB) is altered during EAE has not been reported. Here, we demonstrate that endothelial cell proliferation and vessel permeability increase before EAE clinical onset, leading to vascular remodeling and expansion of intestinal villi capillary bed during disease symptomatic phase in an antigen-independent manner. Concomitant to onset of angiogenesis observed prior to neurological symptoms, we identify an increase of intestinal perivascular immune cells characterized by the surface marker lymphatic vessel endothelial hyaluronic acid receptor 1 (LYVE-1). LYVE-1+ is expressed more frequently on B cells that show high levels of CD73 and have proangiogenic properties. B cell depletion was sufficient to mitigate enteric blood endothelial cell proliferation following immunization for EAE. In conclusion, we propose that altered intestinal vasculature driven by a specialized LYVE-1+ B cell subset promotes angiogenesis and that loss of GVB function is implicated in EAE development and autoimmunity.


Asunto(s)
Linfocitos B , Encefalomielitis Autoinmune Experimental , Animales , Ratones , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Eje Cerebro-Intestino/inmunología , Neovascularización Patológica/inmunología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/metabolismo , Modelos Animales de Enfermedad , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/irrigación sanguínea , Intestinos/patología , Ratones Endogámicos C57BL , Proliferación Celular , Femenino , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Angiogénesis
2.
J Toxicol Environ Health B Crit Rev ; 27(7): 233-263, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-38994870

RESUMEN

The microbiome-gut-brain axis is altered by environmental stressors such as heat, diet, and pollutants as well as microbes in the air, water, and soil. These stressors might alter the host's microbiome and symbiotic relationship by modifying the microbial composition or location. Compartmentalized mutualistic microbes promote the beneficial interactions in the host leading to circulating metabolites and hormones such as insulin and leptin that affect inter-organ functions. Inflammation and oxidative stress induced by environmental stressors may alter the composition, distribution, and activities of the microbes in the microbiomes such that the resultant metabolite and hormone changes are no longer beneficial. The microbiome-gut-brain axis and immune adverse changes that may accompany environmental stressors are reviewed for effects on innate and adaptive immune cells, which may make host immunity less responsive to pathogens and more reactive to self-antigens. Cardiovascular and fluid exchanges to organs might adversely alter organ functionality. Organs, especially the brain, need a consistent supply of nutrients and clearance of debris; disruption of these exchanges by stressors, and involvement of gut microbiome are discussed regarding neural dysfunctions with Alzheimer's disease, autistic spectrum disorders, viral infections, and autoimmune diseases. The focus of this review includes the manner in which environmental stressors may disrupt gut microbiota leading to adverse immune and hormonal influences on development of neuropathology related to hyperhomocysteinemia, inflammation, and oxidative stress, and how certain therapeutics may be beneficial. Strategies are explored to lessen detrimental effects of environmental stressors on central and peripheral health navigated toward (1) understanding neurological disorders and (2) promoting environmental and public health and well-being.


Asunto(s)
Eje Cerebro-Intestino , Microbioma Gastrointestinal , Enfermedades del Sistema Nervioso , Humanos , Microbioma Gastrointestinal/inmunología , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/inmunología , Enfermedades del Sistema Nervioso/inmunología , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/microbiología , Animales , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/toxicidad , Encéfalo/inmunología
3.
Cell Immunol ; 401-402: 104844, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38901288

RESUMEN

The gastrointestinal (GI) tract and the brain form bidirectional nervous, immune, and endocrine communications known as the gut-brain axis. Several factors can affect this axis; among them, various studies have focused on the microbiota and imply that alterations in microbiota combinations can influence both the brain and GI. Also, many studies have shown that the immune system has a vital role in varying gut microbiota combinations. In the current paper, we will review the multidirectional effects of gut microbiota, immune system, and nervous system on each other. Specifically, this review mainly focuses on the impact of Peyer's patches as a critical component of the gut immune system on the gut-brain axis through affecting the gut's microbial composition. In this way, some factors were discussed as proposed elements of missing gaps in this field.


Asunto(s)
Eje Cerebro-Intestino , Microbioma Gastrointestinal , Ganglios Linfáticos Agregados , Ganglios Linfáticos Agregados/inmunología , Humanos , Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Animales , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/inmunología , Encéfalo/inmunología , Encéfalo/fisiología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/inmunología
4.
Front Immunol ; 15: 1401967, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38915399

RESUMEN

Glioblastoma (GBM) is a highly malignant, invasive, and poorly prognosed brain tumor. Unfortunately, active comprehensive treatment does not significantly prolong patient survival. With the deepening of research, it has been found that gut microbiota plays a certain role in GBM, and can directly or indirectly affect the efficacy of immune checkpoint inhibitors (ICIs) in various ways. (1) The metabolites produced by gut microbiota directly affect the host's immune homeostasis, and these metabolites can affect the function and distribution of immune cells, promote or inhibit inflammatory responses, affect the phenotype, angiogenesis, inflammatory response, and immune cell infiltration of GBM cells, thereby affecting the effectiveness of ICIs. (2) Some members of the gut microbiota may reverse T cell function inhibition, increase T cell anti-tumor activity, and ultimately improve the efficacy of ICIs by targeting specific immunosuppressive metabolites and cytokines. (3) Some members of the gut microbiota directly participate in the metabolic process of drugs, which can degrade, transform, or produce metabolites, affecting the effective concentration and bioavailability of drugs. Optimizing the structure of the gut microbiota may help improve the efficacy of ICIs. (4) The gut microbiota can also regulate immune cell function and inflammatory status in the brain through gut brain axis communication, indirectly affecting the progression of GBM and the therapeutic response to ICIs. (5) Given the importance of gut microbiota for ICI therapy, researchers have begun exploring the use of fecal microbiota transplantation (FMT) to transplant healthy or optimized gut microbiota to GBM patients, in order to improve their immune status and enhance their response to ICI therapy. Preliminary studies suggest that FMT may enhance the efficacy of ICI therapy in some patients. In summary, gut microbiota plays a crucial role in regulating ICIs in GBM, and with a deeper understanding of the relationship between gut microbiota and tumor immunity, it is expected to develop more precise and effective personalized ICI therapy strategies for GBM, in order to improve patient prognosis.


Asunto(s)
Neoplasias Encefálicas , Microbioma Gastrointestinal , Glioblastoma , Inhibidores de Puntos de Control Inmunológico , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/inmunología , Glioblastoma/inmunología , Glioblastoma/tratamiento farmacológico , Glioblastoma/terapia , Glioblastoma/microbiología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/microbiología , Animales , Eje Cerebro-Intestino/inmunología , Trasplante de Microbiota Fecal , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos
5.
Front Immunol ; 15: 1365871, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38756771

RESUMEN

More than 20% of American adults live with a mental disorder, many of whom are treatment resistant or continue to experience symptoms. Other approaches are needed to improve mental health care, including prevention. The role of the microbiome has emerged as a central tenet in mental and physical health and their interconnectedness (well-being). Under normal conditions, a healthy microbiome promotes homeostasis within the host by maintaining intestinal and brain barrier integrity, thereby facilitating host well-being. Owing to the multidirectional crosstalk between the microbiome and neuro-endocrine-immune systems, dysbiosis within the microbiome is a main driver of immune-mediated systemic and neural inflammation that can promote disease progression and is detrimental to well-being broadly and mental health in particular. In predisposed individuals, immune dysregulation can shift to autoimmunity, especially in the presence of physical or psychological triggers. The chronic stress response involves the immune system, which is intimately involved with the gut microbiome, particularly in the process of immune education. This interconnection forms the microbiota-gut-immune-brain axis and promotes mental health or disorders. In this brief review, we aim to highlight the relationships between stress, mental health, and the gut microbiome, along with the ways in which dysbiosis and a dysregulated immune system can shift to an autoimmune response with concomitant neuropsychological consequences in the context of the microbiota-gut-immune-brain axis. Finally, we aim to review evidenced-based prevention strategies and potential therapeutic targets.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Disbiosis , Microbioma Gastrointestinal , Trastornos Mentales , Salud Mental , Estrés Psicológico , Humanos , Microbioma Gastrointestinal/inmunología , Eje Cerebro-Intestino/inmunología , Estrés Psicológico/inmunología , Estrés Psicológico/microbiología , Disbiosis/inmunología , Trastornos Mentales/inmunología , Trastornos Mentales/microbiología , Encéfalo/inmunología , Animales , Neuroinmunomodulación
6.
Front Immunol ; 15: 1365673, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38817603

RESUMEN

Importance: Research is beginning to elucidate the sophisticated mechanisms underlying the microbiota-gut-brain-immune interface, moving from primarily animal models to human studies. Findings support the dynamic relationships between the gut microbiota as an ecosystem (microbiome) within an ecosystem (host) and its intersection with the host immune and nervous systems. Adding this to the effects on epigenetic regulation of gene expression further complicates and strengthens the response. At the heart is inflammation, which manifests in a variety of pathologies including neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis (MS). Observations: Generally, the research to date is limited and has focused on bacteria, likely due to the simplicity and cost-effectiveness of 16s rRNA sequencing, despite its lower resolution and inability to determine functional ability/alterations. However, this omits all other microbiota including fungi, viruses, and phages, which are emerging as key members of the human microbiome. Much of the research has been done in pre-clinical models and/or in small human studies in more developed parts of the world. The relationships observed are promising but cannot be considered reliable or generalizable at this time. Specifically, causal relationships cannot be determined currently. More research has been done in Alzheimer's disease, followed by Parkinson's disease, and then little in MS. The data for MS is encouraging despite this. Conclusions and relevance: While the research is still nascent, the microbiota-gut-brain-immune interface may be a missing link, which has hampered our progress on understanding, let alone preventing, managing, or putting into remission neurodegenerative diseases. Relationships must first be established in humans, as animal models have been shown to poorly translate to complex human physiology and environments, especially when investigating the human gut microbiome and its relationships where animal models are often overly simplistic. Only then can robust research be conducted in humans and using mechanistic model systems.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Microbioma Gastrointestinal , Enfermedades Neuroinflamatorias , Humanos , Microbioma Gastrointestinal/inmunología , Animales , Eje Cerebro-Intestino/inmunología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/microbiología , Enfermedades Neuroinflamatorias/etiología , Encéfalo/inmunología , Encéfalo/microbiología
7.
Sci Rep ; 12(1): 1448, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35087123

RESUMEN

Although the etiology of obsessive-compulsive disorder (OCD) is largely unknown, it is accepted that OCD is a complex disorder. There is a known bi-directional interaction between the gut microbiome and brain activity. Several authors have reported associations between changes in gut microbiota and neuropsychiatric disorders, including depression or autism. Furthermore, a pediatric-onset neuropsychiatric OCD-related syndrome occurs after streptococcal infection, which might indicate that exposure to certain microbes could be involved in OCD susceptibility. However, only one study has investigated the microbiome of OCD patients to date. We performed 16S ribosomal RNA gene-based metagenomic sequencing to analyze the stool and oropharyngeal microbiome composition of 32 OCD cases and 32 age and gender matched controls. We estimated different α- and ß-diversity measures and performed LEfSe and Wilcoxon tests to assess differences in bacterial distribution. OCD stool samples showed a trend towards lower bacterial α-diversity, as well as an increase of the relative abundance of Rikenellaceae, particularly of the genus Alistipes, and lower relative abundance of Prevotellaceae, and two genera within the Lachnospiraceae: Agathobacer and Coprococcus. However, we did not observe a different Bacteroidetes to Firmicutes ratio between OCD cases and controls. Analysis of the oropharyngeal microbiome composition showed a lower Fusobacteria to Actinobacteria ratio in OCD cases. In conclusion, we observed an imbalance in the gut and oropharyngeal microbiomes of OCD cases, including, in stool, an increase of bacteria from the Rikenellaceae family, associated with gut inflammation, and a decrease of bacteria from the Coprococcus genus, associated with DOPAC synthesis.


Asunto(s)
Eje Cerebro-Intestino/inmunología , Microbioma Gastrointestinal/inmunología , Trastorno Obsesivo Compulsivo/microbiología , Orofaringe/microbiología , Adulto , Estudios de Casos y Controles , ADN Bacteriano/aislamiento & purificación , Heces/microbiología , Femenino , Microbioma Gastrointestinal/genética , Voluntarios Sanos , Humanos , Masculino , Metagenoma , Metagenómica , Persona de Mediana Edad , Trastorno Obsesivo Compulsivo/inmunología , ARN Ribosómico 16S/genética
8.
Science ; 374(6571): 1087-1092, 2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34822299

RESUMEN

The brain and gastrointestinal tract are critical sensory organs responsible for detecting, relaying, integrating, and responding to signals derived from the internal and external environment. At the interface of this sensory function, immune cells in the intestines and brain consistently survey environmental factors, eliciting responses that inform on the physiological state of the body. Recent research reveals that cross-talk along the gut-brain axis regulates inflammatory nociception, inflammatory responses, and immune homeostasis. Here, we discuss molecular and cellular mechanisms involved in the signaling of inflammation across the gut-brain axis. We further highlight interactions between the gut and the brain in inflammation-associated diseases.


Asunto(s)
Eje Cerebro-Intestino/fisiología , Encéfalo/fisiología , Tracto Gastrointestinal/fisiología , Inflamación/fisiopatología , Transducción de Señal , Animales , Encéfalo/inmunología , Eje Cerebro-Intestino/inmunología , Tracto Gastrointestinal/inmunología , Humanos , Inflamación/inmunología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/fisiología , Nocicepción
9.
Front Immunol ; 12: 742449, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34707612

RESUMEN

The gut-brain axis refers to the bidirectional communication between the gut and brain, and regulates intestinal homeostasis and the central nervous system via neural networks and neuroendocrine, immune, and inflammatory pathways. The development of sequencing technology has evidenced the key regulatory role of the gut microbiota in several neurological disorders, including Parkinson's disease, Alzheimer's disease, and multiple sclerosis. Epilepsy is a complex disease with multiple risk factors that affect more than 50 million people worldwide; nearly 30% of patients with epilepsy cannot be controlled with drugs. Interestingly, patients with inflammatory bowel disease are more susceptible to epilepsy, and a ketogenic diet is an effective treatment for patients with intractable epilepsy. Based on these clinical facts, the role of the microbiome and the gut-brain axis in epilepsy cannot be ignored. In this review, we discuss the relationship between the gut microbiota and epilepsy, summarize the possible pathogenic mechanisms of epilepsy from the perspective of the microbiota gut-brain axis, and discuss novel therapies targeting the gut microbiota. A better understanding of the role of the microbiota in the gut-brain axis, especially the intestinal one, would help investigate the mechanism, diagnosis, prognosis evaluation, and treatment of intractable epilepsy.


Asunto(s)
Eje Cerebro-Intestino/inmunología , Encéfalo/inmunología , Epilepsia/inmunología , Microbioma Gastrointestinal/inmunología , Neuroinmunomodulación/inmunología , Animales , Humanos
10.
Front Immunol ; 12: 718220, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34621267

RESUMEN

A large and expending body of evidence indicates that the gut-brain axis likely plays a crucial role in neurological diseases, including multiple sclerosis (MS). As a whole, the gut-brain axis can be considered as a bi-directional multi-crosstalk pathway that governs the interaction between the gut microbiota and the organism. Perturbation in the commensal microbial population, referred to as dysbiosis, is frequently associated with an increased intestinal permeability, or "leaky gut", which allows the entrance of exogeneous molecules, in particular bacterial products and metabolites, that can disrupt tissue homeostasis and induce inflammation, promoting both local and systemic immune responses. An altered gut microbiota could therefore have significant repercussions not only on immune responses in the gut but also in distal effector immune sites such as the CNS. Indeed, the dysregulation of this bi-directional communication as a consequence of dysbiosis has been implicated as playing a possible role in the pathogenesis of neurological diseases. In multiple sclerosis (MS), the gut-brain axis is increasingly being considered as playing a crucial role in its pathogenesis, with a major focus on specific gut microbiota alterations associated with the disease. In both MS and its purported murine model, experimental autoimmune encephalomyelitis (EAE), gastrointestinal symptoms and/or an altered gut microbiota have been reported together with increased intestinal permeability. In both EAE and MS, specific components of the microbiota have been shown to modulate both effector and regulatory T-cell responses and therefore disease progression, and EAE experiments with germ-free and specific pathogen-free mice transferred with microbiota associated or not with disease have clearly demonstrated the possible role of the microbiota in disease pathogenesis and/or progression. Here, we review the evidence that can point to two possible consequences of the gut-brain axis dysfunction in MS and EAE: 1. A pro-inflammatory intestinal environment and "leaky" gut induced by dysbiosis could lead to an altered communication with the CNS through the cholinergic afferent fibers, thereby contributing to CNS inflammation and disease pathogenesis; and 2. Neuroinflammation affecting efferent cholinergic transmission could result in intestinal inflammation as disease progresses.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo/metabolismo , Susceptibilidad a Enfermedades , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Animales , Biomarcadores , Encéfalo/inmunología , Encéfalo/patología , Eje Cerebro-Intestino/inmunología , Comunicación Celular , Terapia Combinada/métodos , Manejo de la Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Microbioma Gastrointestinal/inmunología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/inervación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia
11.
J Alzheimers Dis ; 84(2): 479-490, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34569965

RESUMEN

Accumulating evidence show that the gut microbiota is deeply involved not only in host nutrient metabolism but also in immune function, endocrine regulation, and chronic disease. In neurodegenerative conditions such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis, the gut-brain axis, the bidirectional interaction between the brain and the gut, provides new route of pathological spread and potential therapeutic targets. Although studies of gut microbiota have been conducted mainly in mice, mammalian gut microbiota is highly diverse, complex, and sensitive to environmental changes. Drosophila melanogaster, a fruit fly, has many advantages as a laboratory animal: short life cycle, numerous and genetically homogenous offspring, less ethical concerns, availability of many genetic models, and low maintenance costs. Drosophila has a simpler gut microbiota than mammals and can be made to remain sterile or to have standardized gut microbiota by simple established methods. Research on the microbiota of Drosophila has revealed new molecules that regulate the brain-gut axis, and it has been shown that dysbiosis of the fly microbiota worsens lifespan, motor function, and neurodegeneration in AD and PD models. The results shown in fly studies represents a fundamental part of the immune and proteomic process involving gut-microbiota interactions that are highly conserved. Even though the fly's gut microbiota are not simple mimics of humans, flies are a valuable system to learn the molecular mechanisms of how the gut microbiota affect host health and behavior.


Asunto(s)
Eje Cerebro-Intestino/inmunología , Drosophila melanogaster , Tracto Gastrointestinal , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/metabolismo , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Drosophila melanogaster/metabolismo , Drosophila melanogaster/microbiología , Disbiosis , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Humanos , Longevidad , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/metabolismo , Proteómica
12.
J Neuroimmunol ; 360: 577700, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34482269

RESUMEN

Gut microbiota composition may affect the central nervous system (CNS) and immune function. Several studies have recently examined the possible link between gut microbiota composition and multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Most of these studies agree that patients with MS suffer from dysbiosis. Moreover, an altered proportion of certain phyla of bacteria was detected in the digestive tracts of these patients compared to healthy individuals. This review article gathers information from research papers that have examined the relationship between gut microbiota composition and MS and its possible mechanisms.


Asunto(s)
Eje Cerebro-Intestino , Disbiosis/complicaciones , Encefalomielitis Autoinmune Experimental/microbiología , Microbioma Gastrointestinal , Esclerosis Múltiple/microbiología , Animales , Eje Cerebro-Intestino/inmunología , Eje Cerebro-Intestino/fisiología , Modelos Animales de Enfermedad , Disbiosis/fisiopatología , Disbiosis/terapia , Encefalomielitis Autoinmune Experimental/fisiopatología , Trasplante de Microbiota Fecal , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/terapia , Enfermedades Neurodegenerativas/etiología , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/microbiología , Probióticos , Ratas , Vitamina D/uso terapéutico
13.
Immunol Lett ; 238: 40-46, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34320384

RESUMEN

Sepsis is a life-threatening condition characterized by an acute cytokine storm followed by prolonged dysfunction of the immune system in the survivors. Post-septic lymphopenia and functional deficits of the remaining immune cells lead to increased susceptibility to secondary infections and other morbid conditions causing late death in the patients. This state of post-septic immunoparalysis may also influence disorders stemming from inappropriate or overactive immune responses, such as autoimmune and immunoinflammatory diseases, including multiple sclerosis. In addition, ongoing autoimmunity likely influences the susceptibility to and outcome of sepsis. This review article addresses the bidirectional relationship between sepsis and multiple sclerosis, with a focus on the immunologic mechanisms of the interaction and potential directions for future studies.


Asunto(s)
Susceptibilidad a Enfermedades , Esclerosis Múltiple/etiología , Sepsis/etiología , Animales , Biomarcadores , Eje Cerebro-Intestino/inmunología , Susceptibilidad a Enfermedades/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/metabolismo , Neuroinmunomodulación , Especificidad de Órganos/inmunología , Sepsis/diagnóstico , Sepsis/metabolismo
14.
Neuropharmacology ; 195: 108682, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34175326

RESUMEN

The vagus nerve is one of the major signalling components between the gut microbiota and brain. However, the exact relationship between gut-brain signaling along the vagus and the effects of gut microbes on brain function and behaviour is unclear. In particular, the relationship between the vagus nerve and immune signaling, that also appears to play a critical role in microbiota-gut-brain communication, has not been delineated. The aim of the present study was to determine the effect of subdiaphragmatic vagotomy on peripheral and central immune changes associated with the anxiolytic actions of L.rhamnosus. Male mice underwent vagotomy or sham surgery, followed by administration of L.rhamnosus for 14 days. L.rhamnosus administration following sham surgery resulted in reduced anxiety-like behaviour, and an attenuation of the hypothalamic-pituitary-adrenal axis (HPA axis), as indicated by reduced plasma corticosterone after acute restraint stress. These effects were associated with an increase in splenic T regulatory cells and a decrease in activated microglia in the hippocampus. The anxiolytic effects, HPA modulation and increase in T regulatory cells were prevented by vagotomy, whereas vagotomy alone led to a significant increase in activated microglia in the hippocampus that was not altered with L.rhamnosus treatment. Thus, both microbe induced and constitutive vagal signaling influences critical immune components of the microbiota-gut-brain axis. These findings suggest that, rather than acting as a direct neural link to the central nervous system, the role of the vagus nerve in gut-microbe to brain signalling is as an integral component of a bi-directional neuroimmunoendocrine pathway.


Asunto(s)
Conducta Animal/efectos de los fármacos , Eje Cerebro-Intestino/efectos de los fármacos , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/diagnóstico por imagen , Lacticaseibacillus rhamnosus , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Animales , Eje Cerebro-Intestino/inmunología , Sistema Hipotálamo-Hipofisario/inmunología , Masculino , Ratones , Sistema Hipófiso-Suprarrenal/inmunología , Vagotomía
15.
Int Immunol ; 33(6): 349-356, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33912906

RESUMEN

The hallmark of inflammatory bowel diseases (IBD) is chronic intestinal inflammation with typical onset in adolescents and young adults. An abundance of neutrophils is seen in the inflammatory lesions, but adaptive immunity is also an important player in the chronicity of the disease. There is an unmet need for new treatment options since modern medicines such as biological therapy with anti-cytokine antibodies still leave a substantial number of patients with persisting disease activity. The role of the central nervous system and its interaction with the gut in the pathophysiology of IBD have been brought to attention both in animal models and in humans after the discovery of the inflammatory reflex. The suggested control of gut immunity by the brain-gut axis represents a novel therapeutic target suitable for bioelectronic intervention. In this review, we discuss the role of the inflammatory reflex in gut inflammation and the recent advances in the treatment of IBD by intervening with the brain-gut axis through bioelectronic devices.


Asunto(s)
Eje Cerebro-Intestino/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Inmunidad Adaptativa/inmunología , Animales , Humanos
16.
Int Immunol ; 33(6): 337-348, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33788920

RESUMEN

Accumulating evidence shows that intestinal homeostasis is mediated by cross-talk between the nervous system, enteric neurons and immune cells, together forming specialized neuroimmune units at distinct anatomical locations within the gut. In this review, we will particularly discuss how the intrinsic and extrinsic neuronal circuitry regulates macrophage function and phenotype in the gut during homeostasis and aberrant inflammation, such as observed in inflammatory bowel disease (IBD). Furthermore, we will provide an overview of basic and translational IBD research using these neuronal circuits as a novel therapeutic tool. Finally, we will highlight the different challenges ahead to make bioelectronic neuromodulation a standard treatment for intestinal immune-mediated diseases.


Asunto(s)
Eje Cerebro-Intestino/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Animales , Homeostasis/inmunología , Humanos , Neuronas/inmunología
17.
Neuroreport ; 32(6): 458-464, 2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33657076

RESUMEN

BACKGROUND: Interest is growing in the role played by intestinal flora in the pathogeneses of diseases and in the possibility of treating disease by altering intestinal flora compositions. Recent studies have focused on the relationship between the intestinal microbiome and brain function as proposed by the brain-gut axis hypothesis. OBJECTIVES: To investigate the relation between ischemic stroke and plasma equol monosulfate levels (a soy isoflavone metabolite) in a middle cerebral artery occlusion (MCAO) mouse model. METHODS: Mice (C57BL/6) were subjected to MCAO for various times (30 min to 24 h), and degrees of cerebral damage were assessed using total infarction volumes, brain edema severities and neurological deficit scores. Hematoxylin and eosin and cresyl violet staining were used to observe morphological changes in ischemic brains. Levels of equol monosulfate in plasma and the relationships between these and degree of brain injury were investigated. RESULTS: Infarction volumes, brain edema severity and neurological deficit scores were significantly correlated with ischemic time, and morphological deteriorations of brain neuronal cells also increased with ischemic duration. Equol monosulfate contents were ischemic-time dependently lower in MCAO treated animals than in sham-operated controls. CONCLUSION: Ischemic stroke may time-dependently reduce plasma levels of equol monosulfate by lowering the metabolic rate of equol in MCAO-induced mice. This study provides indirect support of the brain-gut axis hypothesis.


Asunto(s)
Eje Cerebro-Intestino/fisiología , Equol/sangre , Microbioma Gastrointestinal , Accidente Cerebrovascular Isquémico/sangre , Animales , Edema Encefálico/sangre , Edema Encefálico/inmunología , Edema Encefálico/patología , Edema Encefálico/fisiopatología , Eje Cerebro-Intestino/inmunología , Región CA1 Hipocampal/patología , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Accidente Cerebrovascular Isquémico/inmunología , Accidente Cerebrovascular Isquémico/patología , Accidente Cerebrovascular Isquémico/fisiopatología , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Sulfatos/sangre , Factores de Tiempo
18.
Acta sci., Health sci ; 43: e52932, Feb.11, 2021.
Artículo en Inglés | LILACS | ID: biblio-1368494

RESUMEN

Unravelling the efficacy of gut biome has a major impact on health. An unbalanced microbiome composition is linked to many common illnesses such as gut dysbiosis, mental deformities and immunological imbalance. An optimistic influence on the gut biome can be made by consumingprobiotics. This would stimulate neuroprotection and immunomodulation intended by heavy metals pollution. Lead is a major source of neurotoxin that can induce neural deformities. Lactobacillusspecies isolated from curd were characterized to confirm its specificity. Zebra fish was reared at standard conditions and preclinical assessment on the intensity of induced neurotoxin lead was performed. The embryo toxic assay, immunomodulation effects and animal behavioural models endorsed the consequence of neurotoxicity. Different concentrations of bacterial isolate with standard antidepressant was considered for analysing the vigour of toxicity and its influence on cognitive behaviour by novel tank diving method. The restrain in the animal behaviour was also conferred by all the test samples with a decreased bottom dwelling time which was authenticated with haematology and histopathological studies. The alterations in morphology of the lymphocytes were balanced by the treated test samples. This study paves a twofold potential of probiotic as neuroprotectant and immune modulator against heavy metal toxicity.


Asunto(s)
Animales , Bacterias/patogenicidad , Pez Cebra , Probióticos/análisis , Neuroprotección/inmunología , Eje Cerebro-Intestino/inmunología , Plomo/análisis , Bacterias/virología , Anomalías Congénitas/virología , Linfocitos/microbiología , Metales Pesados/análisis , Toxicidad , Inmunomodulación/inmunología , Disbiosis/microbiología , Lactobacillus/inmunología
19.
Front Immunol ; 12: 828887, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35154087

RESUMEN

Objective: Probiotics can modulate immune responses to resist influenza infection. This study aims to evaluate the anti-viral efficacy of B. dorei. Methods: C57BL/6J mice were infected with influenza virus together with treatment of PBS vehicle, B. dorei, or oseltamivir respectively. Anti-influenza potency of B. dorei and the underlying mechanism were determined by measuring survival rate, lung viral load and pathology, gene expression and production of cytokines and chemokines, and analysis of gut microbiota. Results: Administration of B. dorei increased (by 30%) the survival of influenza-infected mice, and improved their weight loss, lung pathology, lung index, and colon length compared to the vehicle control group. B. dorei treatment reduced (by 61%) the viral load of lung tissue and increased expression of type 1 interferon more rapidly at day 3 postinfection. At day 7 postinfection, B. dorei-treated mice showed lower local (lung) and systemic (serum) levels of interferon and several proinflammatory cytokines or chemokines (IL-1ß, IL-6, TNF-α, IL-10, MCP-1 and IP-10) with a efficacy comparable to oseltamivi treatment. B. dorei treatment also altered gut microbiota as indicated by increased levels of Bacteroides, Prevotella, and Lactobacillus and decreased levels of Escherichia, Shigella, and Parabacteroides. Conclusion: B. dorei has anti-influenza effect. Its working mechanisms involve promoting earlier interferon expression and down-regulating both local and systemic inflammatory response. B. dorei changes the composition of gut microbiota, which may also contribute to its beneficial effects.


Asunto(s)
Bacteroides/inmunología , Interacciones Huésped-Patógeno/inmunología , Virus de la Influenza A/fisiología , Interacciones Microbianas/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Animales , Biomarcadores , Eje Cerebro-Intestino/inmunología , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal/inmunología , Inmunomodulación , Interferones/metabolismo , Metagenoma , Metagenómica/métodos , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Probióticos , Quercetina/metabolismo , Carga Viral
20.
Front Immunol ; 12: 789647, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34992606

RESUMEN

Tetratricopeptide repeat and ankyrin repeat containing 1 (TRANK1) is a robust risk gene of bipolar disorder (BD). However, little is known on the role of TRANK1 in the pathogenesis of BD and whether the gut microbiota is capable of regulating TRANK1 expression. In this study, we first investigated the serum mRNA level of TRANK1 in medication-free patients with a depressive episode of BD, then a mice model was constructed by fecal microbiota transplantation (FMT) to explore the effects of gut microbiota on brain TRANK1 expression and neuroinflammation, which was further verified by in vitro Lipopolysaccharide (LPS) treatment in BV-2 microglial cells and neurons. 22 patients with a depressive episode and 28 healthy individuals were recruited. Serum level of TRANK1 mRNA was higher in depressed patients than that of healthy controls. Mice harboring 'BD microbiota' following FMT presented depression-like phenotype. mRNA levels of inflammatory cytokines and TRANK1 were elevated in mice hippocampus and prefrontal cortex. In vitro, LPS treatment activated the secretion of pro-inflammatory factors in BV-2 cells, which was capable of upregulating the neuronal expression of TRANK1 mRNA. Moreover, primary cortical neurons transfected with plasmid Cytomegalovirus DNA (pcDNA3.1(+)) vector encoding human TRANK1 showed decreased dendritic spine density. Together, these findings add new evidence to the microbiota-gut-brain regulation in BD, indicating that microbiota is possibly involved in the neuropathogenesis of BD by modulating the expression of TRANK1.


Asunto(s)
Trastorno Bipolar/inmunología , Eje Cerebro-Intestino/inmunología , Citocinas/metabolismo , Depresión/inmunología , Microbioma Gastrointestinal/inmunología , Adolescente , Adulto , Animales , Trastorno Bipolar/sangre , Trastorno Bipolar/microbiología , Trastorno Bipolar/patología , Estudios de Casos y Controles , Línea Celular , Citocinas/análisis , Depresión/sangre , Depresión/microbiología , Depresión/patología , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Femenino , Voluntarios Sanos , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Microglía/inmunología , Microglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Corteza Prefrontal/inmunología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Cultivo Primario de Células , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...