Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 781
Filtrar
1.
Rev Esc Enferm USP ; 58: e20230365, 2024.
Artículo en Inglés, Portugués | MEDLINE | ID: mdl-38743953

RESUMEN

OBJECTIVE: To map the evidence in the literature about the relationship between gastrointestinal symptoms and COVID-19 in the pediatric population. METHOD: This is a scoping review following the recommendations of the Joanna Briggs Institute and PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. The search was carried out on the following bases: Embase, Google Scholar, PubMed, Scopus, LILACS, CINAHL, Scielo, Web of Science and Virtual Health Library Portal, between July and August 2023. Original studies available in full, in any language, were included. RESULTS: Ten studies were chosen that pointed to three premises: (1) the ACE2 receptor is found in the epithelial cells of the gastrointestinal tract; (2) gastrointestinal symptoms are mediated by stress and infection is justified by the gut-brain axis; (3) it develops the process of Multisystem Inflammatory Syndrome in children, affecting the gastrointestinal tract. CONCLUSION: The synthesis of evidence provided three assumptions which guide the origin of gastrointestinal symptoms. The identification of gastrointestinal symptoms in children affected by COVID-19 can assist in the clinical approach and management of care and treatments.


Asunto(s)
COVID-19 , Enfermedades Gastrointestinales , Humanos , COVID-19/complicaciones , Enfermedades Gastrointestinales/virología , Enfermedades Gastrointestinales/epidemiología , Niño , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Síndrome de Respuesta Inflamatoria Sistémica/diagnóstico , Eje Cerebro-Intestino/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo
2.
Sci Rep ; 14(1): 10555, 2024 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719902

RESUMEN

Heat stress exposure in intermittent heat waves and subsequent exposure during war theaters pose a clinical challenge that can lead to multi-organ dysfunction and long-term complications in the elderly. Using an aged mouse model and high-throughput sequencing, this study investigated the molecular dynamics of the liver-brain connection during heat stress exposure. Distinctive gene expression patterns induced by periodic heat stress emerged in both brain and liver tissues. An altered transcriptome profile showed heat stress-induced altered acute phase response pathways, causing neural, hepatic, and systemic inflammation and impaired synaptic plasticity. Results also demonstrated that proinflammatory molecules such as S100B, IL-17, IL-33, and neurological disease signaling pathways were upregulated, while protective pathways like aryl hydrocarbon receptor signaling were downregulated. In parallel, Rantes, IRF7, NOD1/2, TREM1, and hepatic injury signaling pathways were upregulated. Furthermore, current research identified Orosomucoid 2 (ORM2) in the liver as one of the mediators of the liver-brain axis due to heat exposure. In conclusion, the transcriptome profiling in elderly heat-stressed mice revealed a coordinated network of liver-brain axis pathways with increased hepatic ORM2 secretion, possibly due to gut inflammation and dysbiosis. The above secretion of ORM2 may impact the brain through a leaky blood-brain barrier, thus emphasizing intricate multi-organ crosstalk.


Asunto(s)
Encéfalo , Perfilación de la Expresión Génica , Hígado , Animales , Ratones , Hígado/metabolismo , Encéfalo/metabolismo , Masculino , Transcriptoma , Eje Cerebro-Intestino , Respuesta al Choque Térmico/genética , Ratones Endogámicos C57BL , Transducción de Señal , Envejecimiento/genética , Envejecimiento/metabolismo
3.
Food Res Int ; 186: 114404, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729686

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder with an unknown etiology. It is associated with various factors and causes great inconvenience to the patient's life. The gut-brain axis (GBA), which serves as a bidirectional information channel for exchanging information between the gut microbiota and the brain, is vital in studying many neurodegenerative diseases. Dietary flavonoids provide anti-inflammatory and antioxidant benefits, as well as regulating the structure and function of the gut microbiota. The occurrence and development of ASD are associated with dysbiosis of the gut microbiota. Modulation of gut microbiota can effectively improve the severity of ASD. This paper reviews the links between gut microbiota, flavonoids, and ASD, focusing on the mechanism of dietary flavonoids in regulating ASD through the GBA.


Asunto(s)
Trastorno del Espectro Autista , Eje Cerebro-Intestino , Flavonoides , Microbioma Gastrointestinal , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Trastorno del Espectro Autista/microbiología , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/dietoterapia , Flavonoides/farmacología , Dieta , Disbiosis , Encéfalo/metabolismo , Animales , Antioxidantes/farmacología
4.
J Neuroinflammation ; 21(1): 124, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730498

RESUMEN

Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.


Asunto(s)
Antiinflamatorios , Lesiones Traumáticas del Encéfalo , Eje Cerebro-Intestino , Humanos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/etiología
5.
Gut Microbes ; 16(1): 2351520, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38717832

RESUMEN

Links between the gut microbiota and human health have been supported throughout numerous studies, such as the development of neurological disease disorders. This link is referred to as the "microbiota-gut-brain axis" and is the focus of an emerging field of research. Microbial-derived metabolites and gut and neuro-immunological metabolites regulate this axis in health and many diseases. Indeed, assessing these signals, whether induced by microbial metabolites or neuro-immune mediators, could significantly increase our knowledge of the microbiota-gut-brain axis. However, this will require the development of appropriate techniques and potential models. Methods for studying the induced signals originating from the microbiota remain crucial in this field. This review discusses the methods and techniques available for studies of microbiota-gut-brain interactions. We highlight several much-debated elements of these methodologies, including the widely used in vivo and in vitro models, their implications, and perspectives in the field based on a systematic review of PubMed. Applications of various animal models (zebrafish, mouse, canine, rat, rabbit) to microbiota-gut-brain axis research with practical examples of in vitro methods and innovative approaches to studying gut-brain communications are highlighted. In particular, we extensively discuss the potential of "organ-on-a-chip" devices and their applications in this field. Overall, this review sheds light on the most widely used models and methods, guiding researchers in the rational choice of strategies for studies of microbiota-gut-brain interactions.


Asunto(s)
Eje Cerebro-Intestino , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped , Animales , Microbioma Gastrointestinal/fisiología , Eje Cerebro-Intestino/fisiología , Humanos , Encéfalo/microbiología , Encéfalo/metabolismo , Encéfalo/fisiología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo , Modelos Animales , Ratones
6.
Nutrients ; 16(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732599

RESUMEN

In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics-probiotics beneficial to mental health via the gut-brain axis-in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.


Asunto(s)
Trastornos Mentales , Probióticos , Ensayos Clínicos Controlados Aleatorios como Asunto , Humanos , Probióticos/uso terapéutico , Femenino , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/terapia , Trastornos del Conocimiento/tratamiento farmacológico , Masculino , Resultado del Tratamiento , Adulto , Eje Cerebro-Intestino/efectos de los fármacos , Persona de Mediana Edad , Microbioma Gastrointestinal/efectos de los fármacos , Lactobacillus , Anciano , Bifidobacterium
7.
Biosens Bioelectron ; 258: 116298, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38701537

RESUMEN

Wireless activation of the enteric nervous system (ENS) in freely moving animals with implantable optogenetic devices offers a unique and exciting opportunity to selectively control gastrointestinal (GI) transit in vivo, including the gut-brain axis. Programmed delivery of light to targeted locations in the GI-tract, however, poses many challenges not encountered within the central nervous system (CNS). We report here the development of a fully implantable, battery-free wireless device specifically designed for optogenetic control of the GI-tract, capable of generating sufficient light over large areas to robustly activate the ENS, potently inducing colonic motility ex vivo and increased propulsion in vivo. Use in in vivo studies reveals unique stimulation patterns that increase expulsion of colonic content, likely mediated in part by activation of an extrinsic brain-gut motor pathway, via pelvic nerves. This technology overcomes major limitations of conventional wireless optogenetic hardware designed for the CNS, providing targeted control of specific neurochemical classes of neurons in the ENS and brain-gut axis, for direct modulation of GI-transit and associated behaviours in freely moving animals.


Asunto(s)
Sistema Nervioso Entérico , Optogenética , Tecnología Inalámbrica , Animales , Optogenética/instrumentación , Sistema Nervioso Entérico/fisiología , Ratones , Tecnología Inalámbrica/instrumentación , Eje Cerebro-Intestino/fisiología , Técnicas Biosensibles/instrumentación , Diseño de Equipo , Encéfalo/fisiología , Ratones Endogámicos C57BL
8.
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
9.
Biol Res ; 57(1): 23, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38705984

RESUMEN

Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats.


Asunto(s)
Ansiedad , Eje Cerebro-Intestino , Dieta Alta en Grasa , Microbioma Gastrointestinal , Animales , Masculino , Dieta Alta en Grasa/efectos adversos , Microbioma Gastrointestinal/fisiología , Ansiedad/microbiología , Eje Cerebro-Intestino/fisiología , Ratas , Ratas Sprague-Dawley , Obesidad/microbiología , Obesidad/psicología , Obesidad/metabolismo , Transducción de Señal/fisiología , Conducta Animal/fisiología
10.
Adv Protein Chem Struct Biol ; 140: 199-248, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38762270

RESUMEN

The human gut microbiota is a complex and dynamic community of microorganisms, that influence metabolic, neurodevelopmental, and immune pathways. Microbial dysbiosis, characterized by changes in microbial diversity and relative abundances, is implicated in the development of various chronic neurological and neurodegenerative disorders. These disorders are marked by the accumulation of pathological protein aggregates, leading to the progressive loss of neurons and behavioural functions. Dysregulations in protein-protein interaction networks and signalling complexes, critical for normal brain function, are common in neurological disorders but challenging to unravel, particularly at the neuron and synapse-specific levels. To advance therapeutic strategies, a deeper understanding of neuropathogenesis, especially during the progressive disease phase, is needed. Biomarkers play a crucial role in identifying disease pathophysiology and monitoring disease progression. Proteomics, a powerful technology, shows promise in accelerating biomarker discovery and aiding in the development of novel treatments. In this chapter, we provide an in-depth overview of how proteomic techniques, utilizing various biofluid samples from patients with neurological conditions and diverse animal models, have contributed valuable insights into the pathogenesis of numerous neurological disorders. We also discuss the current state of research, potential challenges, and future directions in proteomic approaches to unravel neuro-pathological conditions.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Proteómica , Humanos , Disbiosis/metabolismo , Disbiosis/microbiología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/microbiología , Animales , Eje Cerebro-Intestino , Biomarcadores/metabolismo
11.
Cells ; 13(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38727306

RESUMEN

Parkinson's disease (PD) is recognized as the second most prevalent primary chronic neurodegenerative disorder of the central nervous system. Clinically, PD is characterized as a movement disorder, exhibiting an incidence and mortality rate that is increasing faster than any other neurological condition. In recent years, there has been a growing interest concerning the role of the gut microbiota in the etiology and pathophysiology of PD. The establishment of a brain-gut microbiota axis is now real, with evidence denoting a bidirectional communication between the brain and the gut microbiota through metabolic, immune, neuronal, and endocrine mechanisms and pathways. Among these, the vagus nerve represents the most direct form of communication between the brain and the gut. Given the potential interactions between bacteria and drugs, it has been observed that the therapies for PD can have an impact on the composition of the microbiota. Therefore, in the scope of the present review, we will discuss the current understanding of gut microbiota on PD and whether this may be a new paradigm for treating this devastating disease.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Microbioma Gastrointestinal , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/microbiología , Enfermedad de Parkinson/terapia , Encéfalo/microbiología , Encéfalo/patología , Eje Cerebro-Intestino/fisiología , Animales
12.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38731995

RESUMEN

The gut-brain axis is a bidirectional relationship between the microbiota and the brain; genes related to the brain and gut synaptic formation are similar. Research on the causal effects of gut microbiota on human behavior, brain development, and function, as well as the underlying molecular processes, has emerged in recent decades. Probiotics have been shown in several trials to help reduce anxiety and depressive symptoms. Because of this, probiotic combinations have been tested in in vitro models to see whether they might modulate the gut and alleviate depression and anxiety. Therefore, we sought to determine whether a novel formulation might affect the pathways controlling anxiety and depression states and alter gut barrier activities in a 3D model without having harmful side effects. Our findings indicate that B. bifidum novaBBF7 10 mg/mL, B. longum novaBLG2 5 mg/mL, and L. paracasei TJB8 10 mg/mL may influence the intestinal barrier and enhance the synthesis of short-chain fatty acids. Additionally, the probiotics studied did not cause neuronal damage and, in combination, exert a protective effect against the condition of anxiety and depression triggered by L-Glutamate. All these findings show that probiotics can affect gut function to alter the pathways underlying anxiety and depression.


Asunto(s)
Ansiedad , Depresión , Microbioma Gastrointestinal , Probióticos , Ansiedad/terapia , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Eje Cerebro-Intestino , Suplementos Dietéticos
13.
Birth Defects Res ; 116(4): e2310, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38563145

RESUMEN

INTRODUCTION: In this study, we aimed to investigate the inflammatory factors, oxidative stress, and histopathological consequences of the brain-gut axis in male and female rats prenatally exposed to VPA. METHODS: Pregnant Wistar rats were randomly divided into two groups. The animals received saline, and valproic acid (VPA) (600 mg/kg, i.p.) on embryonic day 12.5 (E12.5). All offspring were weaned on postnatal day 21, and the experiments were done in male and female rats on day 60. The brain and intestine tissues were extracted to assess histopathology, inflammation, and oxidative stress. RESULTS: An increase of interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) and a decrease of interleukin-10 (IL-10) were observed in the two sexes and two tissues of the autistic rats. In the VPA-exposed animals, malondialdehyde (MDA) and protein carbonyl (PC) increased in the brain of both sexes and the intestines of only the males. The total antioxidant capacity (TAC), superoxide dismutase (SOD), and catalase (CAT) significantly decreased in both tissues of male and female autistic groups. Histopathological evaluation showed that the %apoptosis of the cortex in the autistic male and female groups was more than in controls whereas this parameter in the CA1 and CA3 was significant only in the male rats. In the intestine, histopathologic changes were seen only in the male autistic animals. CONCLUSION: The inflammatory and antioxidant factors were in line in the brain-gut axis in male and female rats prenatally exposed to VPA. Histopathological consequences were more significant in the VPA-exposed male animals.


Asunto(s)
Trastorno Autístico , Ácido Valproico , Embarazo , Ratas , Masculino , Femenino , Animales , Ácido Valproico/toxicidad , Trastorno Autístico/inducido químicamente , Antioxidantes/metabolismo , Ratas Wistar , Eje Cerebro-Intestino , Estrés Oxidativo , Interleucina-6
14.
J Toxicol Environ Health A ; 87(11): 471-479, 2024 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-38590254

RESUMEN

Cannabidiol (CBD), a natural component extracted from Cannabis sativa L. exerts neuroprotective, antioxidant, and anti-inflammatory effects in Alzheimer's disease (AD), a disease characterized by impaired cognition and accumulation of amyloid-B peptides (Aß). Interactions between the gut and central nervous system (microbiota-gut-brain axis) play a critical role in the pathogenesis of neurodegenerative disorder AD. At present investigations into the mechanisms underlying the neuroprotective action of CBD in AD are not conclusive. The aim of this study was thus to examine the influence of CBD on cognition and involvement of the microbiota-gut-brain axis using a senescence-accelerated mouse prone 8 (SAMP8) model. Data demonstrated that administration of CBD to SAMP8 mice improved cognitive function as evidenced from the Morris water maze test and increased hippocampal activated microglia shift from M1 to M2. In addition, CBD elevated levels of Bacteriodetes associated with a fall in Firmicutes providing morphologically a protective intestinal barrier which subsequently reduced leakage of intestinal toxic metabolites. Further, CBD was found to reduce the levels of hippocampal and colon epithelial cells lipopolysaccharide (LPS), known to be increased in AD leading to impaired gastrointestinal motility, thereby promoting neuroinflammation and subsequent neuronal death. Our findings demonstrated that CBD may be considered a beneficial therapeutic drug to counteract AD-mediated cognitive impairment and restore gut microbial functions associated with the observed neuroprotective mechanisms.


Asunto(s)
Enfermedad de Alzheimer , Cannabidiol , Disfunción Cognitiva , Ratones , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Cannabidiol/farmacología , Cannabidiol/uso terapéutico , Eje Cerebro-Intestino , Cognición , Disfunción Cognitiva/tratamiento farmacológico , Modelos Animales de Enfermedad
15.
Medicine (Baltimore) ; 103(17): e37960, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669388

RESUMEN

Short-chain fatty acids (SCFAs) are, the metabolic byproducts of intestinal microbiota that, are generated through anaerobic fermentation of undigested dietary fibers. SCFAs play a pivotal role in numerous physiological functions within the human body, including maintaining intestinal mucosal health, modulating immune functions, and regulating energy metabolism. In recent years, extensive research evidence has indicated that SCFAs are significantly involved in the onset and progression of Parkinson disease (PD). However, the precise mechanisms remain elusive. This review comprehensively summarizes the progress in understanding how SCFAs impact PD pathogenesis and the underlying mechanisms. Primarily, we delve into the synthesis, metabolism, and signal transduction of SCFAs within the human body. Subsequently, an analysis of SCFA levels in patients with PD is presented. Furthermore, we expound upon the mechanisms through which SCFAs induce inflammatory responses, oxidative stress, abnormal aggregation of alpha-synuclein, and the intricacies of the gut-brain axis. Finally, we provide a critical analysis and explore the potential therapeutic role of SCFAs as promising targets for treating PD.


Asunto(s)
Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/microbiología , Microbioma Gastrointestinal/fisiología , Ácidos Grasos Volátiles/metabolismo , Eje Cerebro-Intestino/fisiología , Estrés Oxidativo/fisiología
16.
Life Sci ; 347: 122650, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38631669

RESUMEN

AIMS: As a unique iron-dependent non-apoptotic cell death, Ferroptosis is involved in the pathogenesis and development of many human diseases and has become a research hotspot in recent years. However, the regulatory role of ferroptosis in the gut-liver-brain axis has not been elucidated. This paper summarizes the regulatory role of ferroptosis and provides theoretical basis for related research. MATERIALS AND METHODS: We searched PubMed, CNKI and Wed of Science databases on ferroptosis mediated gut-liver-brain axis diseases, summarized the regulatory role of ferroptosis on organ axis, and explained the adverse effects of related regulatory effects on various diseases. KEY FINDINGS: According to our summary, the main way in which ferroptosis mediates the gut-liver-brain axis is oxidative stress, and the key cross-talk of ferroptosis affecting signaling pathway network is Nrf2/HO-1. However, there were no specific marker between different organ axes mediate by ferroptosis. SIGNIFICANCE: Our study illustrates the main ways and key cross-talk of ferroptosis mediating the gut-liver-brain axis, providing a basis for future research.


Asunto(s)
Encéfalo , Ferroptosis , Hígado , Estrés Oxidativo , Ferroptosis/fisiología , Humanos , Estrés Oxidativo/fisiología , Encéfalo/metabolismo , Hígado/metabolismo , Hígado/patología , Animales , Eje Cerebro-Intestino/fisiología , Transducción de Señal , Factor 2 Relacionado con NF-E2/metabolismo
17.
CNS Neurosci Ther ; 30(4): e14704, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38584341

RESUMEN

BACKGROUND: The gut microbiome is composed of various microorganisms such as bacteria, fungi, and protozoa, and constitutes an important part of the human gut. Its composition is closely related to human health and disease. Alzheimer's disease (AD) is a neurodegenerative disease whose underlying mechanism has not been fully elucidated. Recent research has shown that there are significant differences in the gut microbiota between AD patients and healthy individuals. Changes in the composition of gut microbiota may lead to the development of harmful factors associated with AD. In addition, the gut microbiota may play a role in the development and progression of AD through the gut-brain axis. However, the exact nature of this relationship has not been fully understood. AIMS: This review will elucidate the types and functions of gut microbiota and their relationship with AD and explore in depth the potential mechanisms of gut microbiota in the occurrence of AD and the prospects for treatment strategies. METHODS: Reviewed literature from PubMed and Web of Science using key terminologies related to AD and the gut microbiome. RESULTS: Research indicates that the gut microbiota can directly or indirectly influence the occurrence and progression of AD through metabolites, endotoxins, and the vagus nerve. DISCUSSION: This review discusses the future challenges and research directions regarding the gut microbiota in AD. CONCLUSION: While many unresolved issues remain regarding the gut microbiota and AD, the feasibility and immense potential of treating AD by modulating the gut microbiota are evident.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Enfermedades Neurodegenerativas , Humanos , Enfermedad de Alzheimer/terapia , Eje Cerebro-Intestino , PubMed , Encéfalo
18.
Neurosci Biobehav Rev ; 161: 105653, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582194

RESUMEN

The evolution of the gut-microbiota-brain axis in animals reveals that microbial inputs influence metabolism, the regulation of inflammation and the development of organs, including the brain. Inflammatory, neurodegenerative and psychiatric disorders are more prevalent in people of low socioeconomic status (SES). Many aspects of low SES reduce exposure to the microbial inputs on which we are in a state of evolved dependence, whereas the lifestyle of wealthy citizens maintains these exposures. This partially explains the health deficit of low SES, so focussing on our evolutionary history and on environmental and lifestyle factors that distort microbial exposures might help to mitigate that deficit. But the human microbiota is complex and we have poor understanding of its functions at the microbial and mechanistic levels, and in the brain. Perhaps its composition is more flexible than the microbiota of animals that have restricted habitats and less diverse diets? These uncertainties are discussed in relation to the encouraging but frustrating results of attempts to treat psychiatric disorders by modulating the microbiota.


Asunto(s)
Evolución Biológica , Microbioma Gastrointestinal , Clase Social , Humanos , Microbioma Gastrointestinal/fisiología , Animales , Eje Cerebro-Intestino/fisiología , Trastornos Mentales/microbiología , Salud Mental , Estatus Socioeconómico Bajo
19.
J Neurosci ; 44(20)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38594069

RESUMEN

The brain bidirectionally communicates with the gut to control food intake and energy balance, which becomes dysregulated in obesity. For example, endocannabinoid (eCB) signaling in the small-intestinal (SI) epithelium is upregulated in diet-induced obese (DIO) mice and promotes overeating by a mechanism that includes inhibiting gut-brain satiation signaling. Upstream neural and molecular mechanism(s) involved in overproduction of orexigenic gut eCBs in DIO, however, are unknown. We tested the hypothesis that overactive parasympathetic signaling at the muscarinic acetylcholine receptors (mAChRs) in the SI increases biosynthesis of the eCB, 2-arachidonoyl-sn-glycerol (2-AG), which drives hyperphagia via local CB1Rs in DIO. Male mice were maintained on a high-fat/high-sucrose Western-style diet for 60 d, then administered several mAChR antagonists 30 min prior to tissue harvest or a food intake test. Levels of 2-AG and the activity of its metabolic enzymes in the SI were quantitated. DIO mice, when compared to those fed a low-fat/no-sucrose diet, displayed increased expression of cFos protein in the dorsal motor nucleus of the vagus, which suggests an increased activity of efferent cholinergic neurotransmission. These mice exhibited elevated levels of 2-AG biosynthesis in the SI, that was reduced to control levels by mAChR antagonists. Moreover, the peripherally restricted mAChR antagonist, methylhomatropine bromide, and the peripherally restricted CB1R antagonist, AM6545, reduced food intake in DIO mice for up to 24 h but had no effect in mice conditionally deficient in SI CB1Rs. These results suggest that hyperactivity at mAChRs in the periphery increases formation of 2-AG in the SI and activates local CB1Rs, which drives hyperphagia in DIO.


Asunto(s)
Dieta Alta en Grasa , Endocannabinoides , Glicéridos , Ratones Endogámicos C57BL , Obesidad , Transducción de Señal , Transmisión Sináptica , Animales , Endocannabinoides/metabolismo , Masculino , Obesidad/metabolismo , Ratones , Transmisión Sináptica/fisiología , Transmisión Sináptica/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Transducción de Señal/fisiología , Glicéridos/metabolismo , Ácidos Araquidónicos/metabolismo , Ingestión de Alimentos/fisiología , Ingestión de Alimentos/efectos de los fármacos , Antagonistas Muscarínicos/farmacología , Receptores Muscarínicos/metabolismo , Eje Cerebro-Intestino/fisiología
20.
J Agric Food Chem ; 72(18): 10406-10419, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38659208

RESUMEN

The impact of leptin resistance on intestinal mucosal barrier integrity, appetite regulation, and hepatic lipid metabolism through the microbiota-gut-brain-liver axis has yet to be determined. Water extract of Phyllanthus emblica L. fruit (WEPE) and its bioactive compound gallic acid (GA) effectively alleviated methylglyoxal (MG)-triggered leptin resistance in vitro. Therefore, this study investigated how WEPE and GA intervention relieve leptin resistance-associated dysfunction in the intestinal mucosa, appetite, and lipid accumulation through the microbiota-gut-brain-liver axis in high-fat diet (HFD)-fed rats. The results showed that WEPE and GA significantly reduced tissues (jejunum, brain, and liver) MG-evoked leptin resistance, malondialdehyde (MDA), proinflammatory cytokines, SOCS3, orexigenic neuropeptides, and lipid accumulation through increasing leptin receptor, tight junction proteins, antimicrobial peptides, anorexigenic neuropeptides, excretion of fecal triglyceride (TG), and short-chain fatty acids (SCFAs) via a positive correlation with the Allobaculum and Bifidobacterium microbiota. These novel findings suggest that WEPE holds the potential as a functional food ingredient for alleviating obesity and its complications.


Asunto(s)
Encéfalo , Dieta Alta en Grasa , Frutas , Microbioma Gastrointestinal , Homeostasis , Leptina , Hígado , Obesidad , Phyllanthus emblica , Extractos Vegetales , Ratas Sprague-Dawley , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratas , Masculino , Obesidad/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/microbiología , Frutas/química , Hígado/metabolismo , Hígado/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Leptina/metabolismo , Extractos Vegetales/farmacología , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Phyllanthus emblica/química , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Apetito/efectos de los fármacos , Eje Cerebro-Intestino/efectos de los fármacos , Bacterias/clasificación , Bacterias/metabolismo , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA