RESUMEN
Alzheimer's disease (AD) is a degenerative disease that causes a progressive decline in memory and thinking skills. Over the past few years, diverse studies have shown that there is no single cause of AD; instead, it has been reported that factors such as genetics, lifestyle, and environment contribute to the pathogenesis of the disease. In this sense, it has been shown that obesity during middle age is one of the most prominent modifiable risk factors for AD. Of the multiple potential mechanisms linking obesity and AD, the gut microbiota (GM) has gained increasing attention in recent years. However, the underlying mechanisms that connect the GM with the process of neurodegeneration remain unclear. Through this narrative review, we present a comprehensive understanding of how alterations in the GM of people with obesity may result in systemic inflammation and affect pathways related to the pathogenesis of AD. We conclude with an analysis of the relationship between the GM and insulin resistance, a risk factor for AD that is highly prevalent in people with obesity. Understanding the crosstalk between obesity, the GM, and the pathogenesis of AD will help to design new strategies aimed at preventing neurodegeneration.
RESUMEN
BACKGROUND: Depressive symptoms during perinatal significantly impact mothers and infants. Emerging evidence suggests a connection between gut microbiota and mood regulation. This study investigated whether depressive symptoms are associated with changes in the gut microbiota of women during the perinatal period. METHOD: A total of 34 pregnant women were screened for depression using the Edinburgh Postnatal Depression Scale (EPDS) and categorized based on symptom severity. Stool samples were collected during the third trimester and at two postpartum timepoints. All samples underwent 16S rRNA gene sequencing and Quantification of Short-Chain Fatty Acids (SCFA) using a gas chromatograph-mass spectrometer (GC-MS). RESULTS: No differences in SCFA concentrations were observed between groups (p>0.05). However, postpartum women with moderate to severe symptoms (MG group) had a significant increase in Enterobacteriaceae abundance compared to the mild and absent group (AL group) (p<0.05). The Bifidobacterium genus increased significantly in both groups over time (p<0.05). The MG group showed a reduction in depressive symptoms during psychiatric treatment (p<0.05). CONCLUSION: These findings suggest a link between gut microbiota and perinatal depressive symptoms. Further research is needed to understand the broader implications for maternal health through microbiome-targeted approaches.
RESUMEN
Stress, unhealthy lifestyle, and sleep disturbance worsen cognitive function in mood disorders, prompting a rise in the development of integrative health approaches. The recent investigations in the gut-brain axis field highlight the strong interplay among microbiota, inflammation, and mental health. Thus, this study aimed to investigate a new nutraceutical formulation comprising prebiotics, minerals, and silymarin's impact on microbiota, inflammation, mood, and sleep quality. The study evaluated the LL1 + silymarin capsule supplementation over 180 days in overweight adults. We analyzed the fecal gut microbiota using partial 16S rRNA sequences, measured cytokine expression via CBA, collected anthropometric data, quality of life, and sleep questionnaire responses, and obtained plasma samples for metabolic and hormonal analysis at baseline (T0) and 180 days (T180) post-supplementation. Our findings revealed significant reshaping in gut microbiota composition at the phylum, genus, and species levels, especially in the butyrate-producer bacteria post-supplementation. These changes in gut microbiota were linked to enhancements in sleep quality, mood perception, cytokine expression, and anthropometric measures which microbiota-derived short-chain fatty acids might enhance. The supplementation tested in this study seems to be able to improve microbiota composition, reflecting anthropometrics and inflammation, as well as sleep quality and mood improvement.
Asunto(s)
Afecto , Eje Cerebro-Intestino , Suplementos Dietéticos , Microbioma Gastrointestinal , Silimarina , Calidad del Sueño , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Proyectos Piloto , Afecto/efectos de los fármacos , Masculino , Femenino , Silimarina/farmacología , Adulto , Eje Cerebro-Intestino/efectos de los fármacos , Persona de Mediana Edad , Calidad de Vida , Heces/microbiología , Cápsulas , Citocinas/metabolismo , Citocinas/sangre , Sobrepeso , Prebióticos/administración & dosificación , ARN Ribosómico 16SRESUMEN
Accumulating evidence suggests that interactions between the brain and gut microbiota significantly impact brain function and mental health. In the present study, we aimed to investigate whether young, healthy adults without psychiatric diagnoses exhibit differences in metabolic stool and microbiota profiles based on depression/anxiety scores and heart rate variability (HRV) parameters. Untargeted nuclear magnetic resonance-based metabolomics was used to identify fecal metabolic profiles. Results were subjected to multivariate analysis through principal component analysis (PCA) and partial least squares discriminant analysis (PLS-DA), and the metabolites were identified through VIP score. Metabolites separating asymptomatic and symptomatic groups were acetate, valine, and glutamate, followed by sugar regions, glutamine, acetone, valerate, and acetoacetate. The main metabolites identified in high vagal tone (HVT) and low vagal tone (LVT) groups were acetate, valerate, and glutamate, followed by propionate and butyrate. In addition to the metabolites identified by the PLS-DA test, significant differences in aspartate, sarcosine, malate, and methionine were observed between the groups. Levels of acetoacetate were higher in both symptomatic and LVT groups. Valerate levels were significantly increased in the symptomatic group, while isovalerate, propionate, glutamate, and acetone levels were significantly increased in the LVT group. Furthermore, distinct abundance between groups was only confirmed for the Firmicutes phylum. Differences between participants with high and low vagal tone suggest that certain metabolites are involved in communication between the vagus nerve and the brain.
RESUMEN
Multiple Sclerosis (MS) is a debilitating disease that severely affects the central nervous system (CNS). Apart from neurological symptoms, it is also characterized by neuropsychiatric comorbidities, such as anxiety and depression. Phosphodiesterase-5 inhibitors (PDE5Is) such as Sildenafil and Tadalafil have been shown to possess antidepressant-like effects, but the mechanisms underpinning such effects are not fully characterized. To address this question, we used the EAE model of MS, behavioral tests, immunofluorescence, immunohistochemistry, western blot, and 16 S rRNA sequencing. Here, we showed that depressive-like behavior in Experimental Autoimmune Encephalomyelitis (EAE) mice is due to neuroinflammation, reduced synaptic plasticity, dysfunction in glutamatergic neurotransmission, glucocorticoid receptor (GR) resistance, increased blood-brain barrier (BBB) permeability, and immune cell infiltration to the CNS, as well as inflammation, increased intestinal permeability, and immune cell infiltration in the distal colon. Furthermore, 16 S rRNA sequencing revealed that behavioral dysfunction in EAE mice is associated with changes in the gut microbiota, such as an increased abundance of Firmicutes and Saccharibacteria and a reduction in Proteobacteria, Parabacteroides, and Desulfovibrio. Moreover, we detected an increased abundance of Erysipelotrichaceae and Desulfovibrionaceae and a reduced abundance of Lactobacillus johnsonii. Surprisingly, we showed that Tadalafil likely exerts antidepressant-like effects by targeting all aforementioned disease aspects. In conclusion, our work demonstrated that anxiety- and depressive-like behavior in EAE is associated with a plethora of neuroimmune and gut microbiota-mediated mechanisms and that Tadalafil exerts antidepressant-like effects probably by targeting these mechanisms. Harnessing the knowledge of these mechanisms of action of Tadalafil is important to pave the way for future clinical trials with depressed patients.
Asunto(s)
Ansiolíticos , Antidepresivos , Eje Cerebro-Intestino , Depresión , Encefalomielitis Autoinmune Experimental , Inhibidores de Fosfodiesterasa 5 , Tadalafilo , Animales , Femenino , Ratones , Ansiolíticos/administración & dosificación , Antidepresivos/administración & dosificación , Autoinmunidad/efectos de los fármacos , Eje Cerebro-Intestino/efectos de los fármacos , Depresión/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Ratones Endogámicos C57BL , Inhibidores de Fosfodiesterasa 5/administración & dosificación , Tadalafilo/administración & dosificaciónRESUMEN
The aging of populations is a global phenomenon that follows a possible increase in the incidence of neurodegenerative diseases. Alzheimer's, Parkinson's, Multiple Sclerosis, Amyotrophic Lateral Sclerosis, and Huntington's diseases are some neurodegenerative disorders that aging could initiate or aggravate. Recent research has indicated that intestinal microbiota dysbiosis can trigger metabolism and brain functioning, contributing to the etiopathogenesis of those neurodegenerative diseases. The intestinal microbiota and its metabolites show significant functions in various aspects, such as the immune system modulation (development and maturation), the maintenance of the intestinal barrier integrity, the modulation of neuromuscular functions in the intestine, and the facilitation of essential metabolic processes for both the microbiota and humans. The primary evidence supporting the connection between intestinal microbiota and its metabolites with neurodegenerative diseases are epidemiological observations and animal models experimentation. This paper reviews up-to-date evidence on the correlation between the microbiota-gut-brain axis and neurodegenerative diseases, with a specially focus on gut metabolites. Dysbiosis can increase inflammatory cytokines and bacterial metabolites, altering intestinal and blood-brain barrier permeability and causing neuroinflammation, thus facilitating the pathogenesis of neurodegenerative diseases. Clinical data supporting this evidence still needs to be improved. Most of the works found are descriptive and associated with the presence of phyla or species of bacteria with neurodegenerative diseases. Despite the limitations of recent research, the potential for elucidating clinical questions that have thus far eluded clarification within prevailing pathophysiological frameworks of health and disease is promising through investigation of the interplay between the host and microbiota.
Asunto(s)
Eje Cerebro-Intestino , Disbiosis , Microbioma Gastrointestinal , Enfermedades Neurodegenerativas , Humanos , Microbioma Gastrointestinal/fisiología , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/microbiología , Disbiosis/metabolismo , Eje Cerebro-Intestino/fisiología , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismoRESUMEN
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/fisiologíaRESUMEN
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 , BifidobacteriumRESUMEN
Evidence shows that the gut microbiome in early life is an essential modulator of physiological processes related to healthy brain development, as well as mental and neurodegenerative disorders. Here, we conduct a systematic review of gut microbiome assessments on infants (both healthy and with conditions that affect brain development) during the first thousand days of life, associated with neurodevelopmental outcomes, with the aim of investigating key microbiome players and mechanisms through which the gut microbiome affects the brain. Bacteroides and Bifidobacterium were associated with non-social fear behavior, duration of orientation, cognitive and motricity development, and neurotypical brain development. Lachnospiraceae, Streptococcus, and Faecalibacterium showed variable levels of influence on behavior and brain development. Few studies described mechanistic insights related to NAD salvage, aspartate and asparagine biosynthesis, methanogenesis, pathways involved in bile acid transformation, short-chain fatty acids production, and microbial virulence genes. Further studies associating species to gene pathways and robustness in data analysis and integration are required to elucidate the functional mechanisms underlying the role of microbiome-gut-brain axis in early brain development.
RESUMEN
The intensification of production systems has resulted in detrimental effects on sow welfare, which can have an adverse influence on their offspring. Considering the relevance of the microbiota-gut-brain axis, probiotics can mitigate such impacts. To investigate the effects of the dietary inclusion of probiotics on the welfare of sows and piglets, 147 multiparous sows were randomly assigned to two groups: a control group or a group supplemented with a multistrain probiotic from the beginning of pregnancy to the end of lactation. The human-animal relationship (HAR), stereotypic behavior, position changes, salivary cortisol, and plasma serotonin levels were assessed in the sows. The piglets' back test behavior and organ weight were analyzed. The probiotic-supplemented sows exhibited a better HAR index (p = 0.017), which indicated reduced aversion towards humans. The frequency of stereotypies was not influenced by the treatments. However, the supplemented sows spent more time standing (p = 0.054) and less time lying down (p = 0.008). The cortisol level of the supplemented sows was 50% lower (p = 0.047) and the serotonin levels were 11% higher (p = 0.034) than control animals. The multistrain piglets were more passive and less resistant (p = 0.076) in the back test. The organ weights were not influenced by treatments. In conclusion, the sows supplemented with probiotics showed less fear and more motivation indicators, while their piglets showed less aggression behaviors.
RESUMEN
Sleep disruption, especially that resulting from obstructive sleep apnea (OSA) - a widely prevalent sleep disorder - can lead to important systemic repercussions. We raise a subject of current interest, namely the possible relationship between sleep in general, OSA, and irritable bowel syndrome (IBS), an intestinal disease that can be made worse by stressful events. The intermittent hypoxia caused by OSA can induce alterations in the gut microbiota, which can lead to the dysregulation of the gut-brain axis and the worsening of IBS. This may be considered to be a circular relationship, with OSA playing a crucial role in the worsening of bowel symptoms, which in turn have a negative effect on sleep. Thus, based on previous evidence, we suggest that improving sleep quality could be a key to disrupting this relationship of IBS aggravation and OSA.
Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , Apnea Obstructiva del Sueño , Humanos , Microbioma Gastrointestinal/fisiología , Eje Cerebro-Intestino , SueñoRESUMEN
The hypothalamic neuropeptides linked to appetite and satiety were investigated in obese mice treated with cotadutide (a dual receptor agonist of glucagon-like peptide 1 (GLP-1R)/Glucagon (GCGR)). Twelve-week-old male C57BL/6 mice were fed a control diet (C group, n = 20) or a high-fat diet (HF group, n = 20) for ten weeks. Each group was further divided, adding cotadutide treatment and forming groups C, CC, HF, and HFC for four additional weeks. The hypothalamic arcuate neurons were labeled by immunofluorescence, and protein expressions (Western blotting) for neuropeptide Y (NPY), proopiomelanocortin (POMC), agouti-related protein (AgRP), and cocaine- and amphetamine-regulated transcript (CART). Cotadutide enhanced POMC and CART neuropeptides and depressed NPY and AGRP neuropeptides. In addition, gene expressions (RT-qPCR) determined that Lepr (leptin receptor) and Calcr (calcitonin receptor) were diminished in HF compared to C but enhanced in CC compared to C and HFC compared to HF. Besides, Socs3 (suppressor of cytokine signaling 3) was decreased in HFC compared to HF, while Sst (somatostatin) was higher in HFC compared to HF; Tac1 (tachykinin 1) and Mc4r (melanocortin-4-receptor) were lower in HF compared to C but increased in HFC compared to HF. Also, Glp1r and Gcgr were higher in HFC compared to HF. In conclusion, the findings are compelling, demonstrating the effects of cotadutide on hypothalamic neuropeptides and hormone receptors of obese mice. Cotadutide modulates energy balance through the gut-brain axis and its associated signaling pathways. The study provides insights into the mechanisms underlying cotadutide's anti-obesity effects and its possible implications for obesity treatment.
Asunto(s)
Glucagón , Neuropéptidos , Péptidos , Ratones , Animales , Masculino , Proteína Relacionada con Agouti , Glucagón/metabolismo , Ratones Obesos , Proopiomelanocortina/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratones Endogámicos C57BL , Neuropéptidos/genética , Hipotálamo/metabolismo , Neuropéptido Y/genética , Péptido 1 Similar al Glucagón/metabolismoRESUMEN
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.
RESUMEN
Os sintomas depressivos durante a gravidez e o período pós-parto (PP) são prevalentes e podem ter implicações profundas para o bem-estar materno e infantil. Evidências emergentes sugerem que a microbiota intestinal pode desempenhar um papel na regulação do humor. Este estudo explora a relação entre a composição da microbiota intestinal e os sintomas depressivos em mulheres grávidas e no pós-parto com diferentes intensidade de sintomas. Foram recrutadas gestantes que faziam acompanhamento nos hospitais HCFMUSP e HU- USP. A partir do preenchimento do questionário de Escala de Edimburgo as participantes foram triadas para os grupos de sintomas ausentes ou leves (AL) e sintomas graves ou moderados (MG). Para a análise de microbiota, as participantes forneceram amostras de fezes em três momentos diferentes. Uma no terceiro trimestre de gestação (G) e duas no período pós-parto. A primeira amostra deste período foi coletada durante a internação do pós-parto (P1), e a segunda durante a consulta de retorno um mês após o parto (P2). A composição da microbiota intestinal foi analisada usando técnicas de sequenciamento de alto rendimento e os ácidos graxos de cadeia curta (AGCC) foram quantificados por cromatografia gasosa acoplada à espectrometria de massas (GC-MS). Análises bioinformáticas e estatísticas foram realizadas utilizando os softwares QIIME 2 (2022.2) e R (4.3.1) para identificar possíveis associações entre a composição da microbiota intestinal e a gravidade dos sintomas depressivos. Os resultados indicam que a familia Enterobacteriacea aparece com maior abundância nas mulheres do grupo MG, especialmente durante o período P1 (p<0,05) e que há uma diminuição significativa (p<0,05) de sintomas depressivos nas participantes do grupo MG desde sua triagem até o fim do acompanhamento do estudo, indicando que conduta terapêutica está sendo eficaz. Apesar de não ter sido estabelecida diferença estatística na abundância relativa da microbiota entre os grupos durante a gestação e nos índices de alfa e beta diversidade entre grupos e entre os períodos, é possivel observar uma tendência de mudança de microbiota ao longo do tratamento com aumento do gênero Bifidobacterium, diminuição da familia Enterobacteraceae e é possivel observar uma aparente correlação inversa entre a diminuição da intensidade de sintomas depressivos e o aumento da abundância dos gêneros Bifidobacterium e Clostridium, além do aumento das concentrações de AGCC. Em conclusão, a composição da microbiota intestinal parece ser influenciada pela gravidade dos sintomas depressivos em mulheres grávidas e no pós-parto. Pesquisas adicionais são necessárias para explorar a relação entre a microbiota intestinal e a depressão perinatal e determinar as implicações clínicas dessas descobertas para a saúde materna e infantil.
Depressive symptoms during pregnancy and the postpartum period (PP) are prevalent and can have profound implications for maternal and infant well-being. Emerging evidence suggests that the gut microbiota may play a role in mood regulation. This study explores the relationship between gut microbiota composition and depressive symptoms in pregnant and postpartum women with different symptom severities. A cohort of pregnant women were recruited from HCFMUSP and HU-USP. Participants completed standardized depression assessment tools and were allocate in groups of absent or mild depressive symptoms (AL) and moderate or severe depressive symptoms (MG) and provided stool samples in three different time periods. One at the third gestation trimester (G) and two at the postpartum period. The first sample from this period was collected during postpartum hospitalization(P1), and the second during the onemonth postpartum follow-up appointment (P2). Their gut microbiota composition was analyzed using high-throughput sequencing techniques and Gas chromatography mass spectrometry (GS-MS) for quantification of short-chain fatty acids (SCFAs). Bioinformatic and statistical analyses were performed using softwares QIIME 2 (2022.2) and R (4.3.1) to identify potential associations between gut microbiota composition and depressive symptom severity. Findings that the Enterobacteriaceae family appears more abundantly in women of the MG group, especially during period P1 (p<0.05), and that there is a significant decrease (p<0.05) in depressive symptoms among the participants of the MG group from their screening to the end of the study follow-up, suggesting that the therapeutic approach is effective. Although no statistical differences in alpha and beta diversity indices were established between groups and across periods, it is possible to observe a trend of microbiota change during the treatment, with an increase in the Bifidobacterium genus, a decrease in the Enterobacteriaceae family, and an apparent inverse correlation between the reduction in the intensity of depressive symptoms and the increased abundance of the Bifidobacterium, Clostridium, and Dorea genera, as well as an increase in the concentrations of SCFAs. In conclusion, composition of gut microbiota appears to be influenced by the severity of depressive symptoms in pregnant and postpartum women. Further research is warranted to explore links between gut microbiota and perinatal depression and to determine the clinical implications of these findings for maternal and infant health
Asunto(s)
Humanos , Femenino , Embarazo , Embarazo , Depresión/patología , Microbioma Gastrointestinal/inmunología , Obstetricia/clasificación , Derivación y Consulta/clasificación , Espectrometría de Masas/métodos , Bifidobacterium/inmunología , Cromatografía de Gases/instrumentación , Hospitales/clasificación , Bienestar del Lactante/clasificación , Cromatografía de Gases y Espectrometría de Masas/métodos , Bienestar Materno/clasificaciónRESUMEN
The gut-brain axis is an essential communication pathway between the central nervous system (CNS) and the gastrointestinal tract. The human microbiota is composed of a diverse and abundant microbial community that compasses more than 100 trillion microorganisms that participate in relevant physiological functions such as host nutrient metabolism, structural integrity, maintenance of the gut mucosal barrier, and immunomodulation. Recent evidence in animal models has been instrumental in demonstrating the possible role of the microbiota in neurodevelopment, neuroinflammation, and behavior. Furthermore, clinical studies suggested that adverse changes in the microbiota can be considered a susceptibility factor for neurological disorders (NDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). In this review, we will discuss evidence describing the role of gut microbes in health and disease as a relevant risk factor in the pathogenesis of neurodegenerative disorders, including AD, PD, HD, and ALS.
Asunto(s)
Enfermedad de Alzheimer , Esclerosis Amiotrófica Lateral , Microbioma Gastrointestinal , Enfermedad de Huntington , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Animales , Humanos , Eje Cerebro-Intestino , Microbioma Gastrointestinal/fisiología , Enfermedades Neurodegenerativas/patología , Sistema Nervioso Central , Enfermedad de Parkinson/patología , Enfermedad de Huntington/patologíaRESUMEN
The neonatal period represents a critical stage for the establishment and development of the gut microbiota, which profoundly influences the future health trajectory of individuals. This review examines the importance of intestinal microbiota in humans and dogs, aiming to elucidate the distinct characteristics and variations in the composition between these two species. In humans, the intestinal microbiota contributes to several crucial physiological processes, including digestion, nutrient absorption, immune system development, and modulation of host metabolism. Dysbiosis, an imbalance or disruption of the gut microbial community, has been linked to various disorders, such as inflammatory bowel disease, obesity, and even neurological conditions. Furthermore, recent research has unveiled the profound influence of the gut-brain axis, emphasizing the bidirectional communication between the gut microbiota and the central nervous system, impacting cognitive function and mental health. Similarly, alterations in the canine intestinal microbiota have been associated with gastrointestinal disorders, including chronic enteropathy, such as inflammatory bowel disease, food allergies, and ulcerative histiocytic colitis. However, our understanding of the intricacies and functional significance of the intestinal microbiota in dogs remains limited. Understanding the complex dynamics of the intestinal microbiota in both humans and dogs is crucial for devising effective strategies to promote health and manage disease. Moreover, exploring the similarities and differences in the gut microbial composition between these two species can facilitate translational research, potentially leading to innovative therapeutic interventions and strategies to enhance the well-being of both humans and dogs.
RESUMEN
Major depressive disorder (MDD) is a complex mood disorder. While much progress has been made in understanding the pathophysiology of MDD, no single mechanism can explain all facets of this disorder. Several studies show that disturbances in biological rhythms can lead to the development of MDD. Indeed, insomnia or hypersomnia are symptoms included in the MDD diagnostic criteria. Clinical studies and meta-analyses showed a strong relationship between MDD and sleep disorders. Sleep disorder and MDD are associated with activation in the hypothalamicpituitary-adrenal (HPA) axis and inflammation. The increase in inflammatory response can activate the kynurenine pathway, decrease serotonin synthesis, and affect other factors involved in the pathophysiology of neuropsychiatric conditions. Moreover, sleep disorders and MDD can change the gut microbiota and alter the microbiota-gut-brain axis. Thus, this review discusses the relationship between MDD, circadian rhythms, and sleep disorders, describing the potential pathophysiological mechanism shared in these conditions. In addition, therapeutic opportunities based on antiinflammatory, antioxidant, HPA axis regulatory, and synapse-modulating actions are raised. For the article search, we used the PubMed database. Both sleep disorders and changes in biological rhythms have a bidirectional relationship with MDD. Although some pathophysiological mechanisms, including inflammation, changes in the gut microbiota, and decreased neuroplasticity, may be involved in the relationship between sleep, circadian rhythms, and MDD, other mechanisms are not yet well understood. Therapeutic opportunities based on anti-inflammatory, antioxidant, HPA regulatory axis, and synapse modulating actions appear to be promising targets in preventing MDD, circadian rhythm disturbances, and sleep disorders.
RESUMEN
Recent research has focused on the link between diet, intestinal microbiota, and the impact of excessive consumption of saturated fatty acids. Saturated fatty acids, found in animal fats, dairy, and processed foods, contribute to dysbiosis, increase intestinal barrier permeability, chronic low-grade inflammation, oxidative stress, and dysfunction of the blood-brain barrier, affecting the central nervous system. High intake of saturated fatty acids is associated with an increased risk of developing Parkinson's disease (PD). Diets low in saturated fats, rich in fibers, promote microbial diversity, improve gut health, and potentially reduce the risk of neurodegenerative diseases like PD.
Asunto(s)
Microbioma Gastrointestinal , Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Animales , Enfermedad de Parkinson/etiología , Microbioma Gastrointestinal/fisiología , Inflamación , Dieta , Ácidos GrasosRESUMEN
Physical activity has been demonstrated to have a significant impact on gut microbial diversity and function. Emerging research has revealed certain aspects of the complex interactions between the gut, exercise, microbiota, and neurodegenerative diseases, suggesting that changes in gut microbial diversity and metabolic function may have an impact on the onset and progression of neurological conditions. This study aimed to review the current literature from several databases until 1 June 2023 (PubMed/MEDLINE, Web of Science, and Google Scholar) on the interplay between the gut, physical exercise, microbiota, and neurodegeneration. We summarized the roles of exercise and gut microbiota on neurodegeneration and identified the ways in which these are all connected. The gut-brain axis is a complex and multifaceted network that has gained considerable attention in recent years. Research indicates that gut microbiota plays vital roles in metabolic shifts during physiological or pathophysiological conditions in neurodegenerative diseases; therefore, they are closely related to maintaining overall health and well-being. Similarly, exercise has shown positive effects on brain health and cognitive function, which may reduce/delay the onset of severe neurological disorders. Exercise has been associated with various neurochemical changes, including alterations in cortisol levels, increased production of endorphins, endocannabinoids like anandamide, as well as higher levels of serotonin and dopamine. These changes have been linked to mood improvements, enhanced sleep quality, better motor control, and cognitive enhancements resulting from exercise-induced effects. However, further clinical research is necessary to evaluate changes in bacteria taxa along with age- and sex-based differences.