RESUMEN
Neurodevelopmental disorders, including autism spectrum disorder (ASD), are defined by core behavioral impairments; however, subsets of individuals display a spectrum of gastrointestinal (GI) abnormalities. We demonstrate GI barrier defects and microbiota alterations in the maternal immune activation (MIA) mouse model that is known to display features of ASD. Oral treatment of MIA offspring with the human commensal Bacteroides fragilis corrects gut permeability, alters microbial composition, and ameliorates defects in communicative, stereotypic, anxiety-like and sensorimotor behaviors. MIA offspring display an altered serum metabolomic profile, and B. fragilis modulates levels of several metabolites. Treating naive mice with a metabolite that is increased by MIA and restored by B. fragilis causes certain behavioral abnormalities, suggesting that gut bacterial effects on the host metabolome impact behavior. Taken together, these findings support a gut-microbiome-brain connection in a mouse model of ASD and identify a potential probiotic therapy for GI and particular behavioral symptoms in human neurodevelopmental disorders.
Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/microbiología , Tracto Gastrointestinal/microbiología , Animales , Ansiedad/metabolismo , Ansiedad/microbiología , Bacteroides fragilis , Conducta Animal , Encéfalo/fisiología , Niño , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Modelos Animales de Enfermedad , Femenino , Tracto Gastrointestinal/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Microbiota , Probióticos/administración & dosificaciónRESUMEN
Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.
Asunto(s)
Conducta Animal , Vida Libre de Gérmenes , Serotonina , Animales , Serotonina/metabolismo , Ratones , Masculino , Microbioma Gastrointestinal/fisiología , Encéfalo/metabolismo , Triptófano Hidroxilasa/metabolismo , Triptófano Hidroxilasa/genética , Ansiedad/metabolismo , Ansiedad/microbiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Ratones Endogámicos C57BL , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT1A/genética , Colon/metabolismo , Colon/microbiologíaRESUMEN
Emerging evidence suggests that the gut microbiota is closely related to psychiatric disorders. However, little is known about the role of the gut microbiota in the development of obsessive-compulsive disorder (OCD). Here, to investigate the contribution of gut microbiota to the pathogenesis of OCD, we transplanted fecal microbiota from first-episode, drug-naive OCD patients or demographically matched healthy individuals into antibiotic-treated specific pathogen-free (SPF) mice and showed that colonization with OCD microbiota is sufficient to induce core behavioral deficits, including abnormal anxiety-like and compulsive-like behaviors. The fecal microbiota was analyzed using 16 S rRNA full-length sequencing, and the results demonstrated a clear separation of the fecal microbiota of mice colonized with OCD and control microbiota. Notably, microbiota from OCD-colonized mice resulted in injured neuronal morphology and function in the mPFC, with inflammation in the mPFC and colon. Unbiased metabolomic analyses of the serum and mPFC region revealed the accumulation of succinic acid (SA) in OCD-colonized mice. SA impeded neuronal activity and induced an inflammatory response in both the colon and mPFC, impacting intestinal permeability and brain function, which act as vital signal mediators in gut microbiota-brain-immune crosstalk. Manipulations of dimethyl malonate (DM) have been reported to exert neuroprotective effects by suppressing the oxidation of accumulated succinic acid, attenuating the downstream inflammatory response and neuronal damage, and can help to partly improve abnormal behavior and reduce neuroinflammation and intestinal inflammation in OCD-colonized mice. We propose that the gut microbiota likely regulates brain function and behaviors in mice via succinic acid signaling, which contributes to the pathophysiology of OCD through gut-brain crosstalk and may provide new insights into the treatment of this disorder.
Asunto(s)
Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Trastorno Obsesivo Compulsivo , Ácido Succínico , Animales , Trastorno Obsesivo Compulsivo/metabolismo , Trastorno Obsesivo Compulsivo/microbiología , Microbioma Gastrointestinal/fisiología , Ratones , Humanos , Masculino , Ácido Succínico/metabolismo , Trasplante de Microbiota Fecal/métodos , Femenino , Enfermedades Neuroinflamatorias/metabolismo , Heces/microbiología , Adulto , Síntomas Conductuales/metabolismo , Inflamación/metabolismo , Corteza Prefrontal/metabolismo , Ansiedad/metabolismo , Ansiedad/microbiología , Ratones Endogámicos C57BL , Conducta Animal , Encéfalo/metabolismo , Eje Cerebro-Intestino/fisiologíaRESUMEN
OBJECTIVE: The purpose of this study is to investigate the connection of pre-competition anxiety with gut microbiota and metabolites in wrestlers with different sports performances. METHODS: One week prior to a national competition, 12 wrestlers completed anxiety questionnaires. Faecal and urine samples were collected for the analysis of gut microbiota and metabolites through the high-throughput sequencing of the 16 S rRNA gene in conjunction with untargeted metabolomics technology. The subjects were divided into two groups, namely, achievement (CP) and no-achievement (CnP) wrestlers, on the basis of whether or not their performances placed them in the top 16 at the competition. The relationship amongst the variations in gut microbiota, metabolites, and anxiety indicators was analyzed. RESULTS: (1) The CP group exhibited significantly higher levels of "state self-confidence," "self-confidence," and "somatic state anxiety" than the CnP group. Conversely, the CP group displayed lower levels of "individual failure anxiety" and "sports competition anxiety" than the CnP group. (2) The gut microbiota in the CP group was more diverse and abundant than that in the CnP group. Pre-competition anxiety was linked to Oscillospiraceae UCG_005, Paraprevotella, Ruminococcaceae and TM7x. (3) The functions of differential metabolites in faeces and urine of the CP/CnP group were mainly enriched in caffeine metabolism, lipopolysaccharide biosynthesis and VEGF and mTOR signaling pathways. Common differential metabolites in feces and urine were significantly associated with multiple anxiety indicators. CONCLUSIONS: Wrestlers with different sports performance have different pre-competition anxiety states, gut microbiota distribution and abundance and differential metabolites in faeces and urine. A certain correlation exists between these psychological and physiological indicators.
Asunto(s)
Ansiedad , Eje Cerebro-Intestino , Heces , Microbioma Gastrointestinal , Lucha , Microbioma Gastrointestinal/fisiología , Humanos , Ansiedad/microbiología , Masculino , Heces/microbiología , Adulto Joven , Eje Cerebro-Intestino/fisiología , ARN Ribosómico 16S/genética , Bacterias/clasificación , Bacterias/genética , Bacterias/metabolismo , Bacterias/aislamiento & purificación , Adolescente , Metabolómica/métodos , Rendimiento Atlético/fisiología , AdultoRESUMEN
The developing central nervous system is highly sensitive to nutrient changes during the perinatal period, emphasising the potential impact of alterations of maternal diet on offspring brain development and behaviour. A growing body of research implicates the gut microbiota in neurodevelopment and behaviour. Maternal overweight and obesity during the perinatal period has been linked to changes in neurodevelopment, plasticity and affective disorders in the offspring, with implications for microbial signals from the maternal gut. Here we investigate the impact of maternal high-fat diet (mHFD)-induced changes in microbial signals on offspring brain development, and neuroimmune signals, and the enduring effects on behaviour into adolescence. We first demonstrate that maternal caecal microbiota composition at term pregnancy (embryonic day 18: E18) differs significantly in response to maternal diet. Moreover, mHFD resulted in the upregulation of microbial genes in the maternal intestinal tissue linked to alterations in quinolinic acid synthesis and elevated kynurenine levels in the maternal plasma, both neuronal plasticity mediators related to glutamate metabolism. Metabolomics of mHFD embryonic brains at E18 also detected molecules linked to glutamate-glutamine cycle, including glutamic acid, glutathione disulphide, and kynurenine. During adolescence, the mHFD offspring exhibited increased locomotor activity and anxiety-like behaviour in a sex-dependent manner, along with upregulation of glutamate-related genes compared to controls. Overall, our results demonstrate that maternal exposure to high-fat diet results in microbiota changes, behavioural imprinting, altered brain metabolism, and glutamate signalling during critical developmental windows during the perinatal period.
Asunto(s)
Encéfalo , Dieta Alta en Grasa , Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Embarazo , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología , Masculino , Conducta Animal/fisiología , Fenómenos Fisiologicos Nutricionales Maternos , Conducta del Adolescente/fisiología , Ratones , Ansiedad/metabolismo , Ansiedad/microbiologíaRESUMEN
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
Objective: To explore intestinal flora differences in species diversity, community structure, and abundance of breast cancer and non-breast cancer populations with anxiety and depression and the corresponding group without anxiety and depression by 16S rRNA high-throughput sequencing technology. Method: Breast cancer and non-breast cancer participants were recruited based on the inclusion and exclusion criteria as the research subjects. The study employed the anxiety self-assessment scale and the depression self-rating scale in the questionnaire survey to collect data. Results: The scores of anxiety and depression of the four groups are as follows: In the breast cancer with anxiety and/or depression (BCAD) group, the anxiety score is 58.80 ± 5.27 and the depression score is 59.60 ± 4.94. In the breast cancer without anxiety and/or depression (BCWAD) group, the anxiety score is 36.53 ± 4.52 and the depression score is 38.20 ± 3.78. In the non-breast cancer group with anxiety and/or depression (HAD) group, the anxiety score is 57.87 ± 4.53 and the depression score is 59.13 ± 5.24. In the non-breast cancer group without anxiety and depression (HWAD) group, the anxiety score is 35.13 ± 5.28 and the depression score is 32.33 ± 4.37. Conclusion: The intestinal flora of the breast cancer patients is significantly different from those of non-breast cancer patients, suggesting that there is an internal relationship between the changes in the intestinal flora and the occurrence and development of breast cancer. People with anxiety and depression without breast cancer show changes in their intestinal flora, suggesting that the changes of the intestinal flora can indeed trigger anxiety and depression. For the breast cancer patients with anxiety and depression, the intestinal flora shows a decrease in diversity and abundance, suggesting that the intestinal flora of the breast cancer patients with anxiety and depression undergo further changes. Thus the intestinal flora can become a new tool for monitoring, preventing, and treating the breast cancer and negative emotions.
Asunto(s)
Ansiedad , Neoplasias de la Mama , Depresión , Microbioma Gastrointestinal , ARN Ribosómico 16S , Humanos , Neoplasias de la Mama/psicología , Neoplasias de la Mama/microbiología , Femenino , Microbioma Gastrointestinal/genética , Ansiedad/microbiología , Depresión/microbiología , Persona de Mediana Edad , ARN Ribosómico 16S/genética , Adulto , AncianoRESUMEN
Migraine is a common and debilitating neurological disorder characterized by the recurrent attack of pulsating headaches typically localized on one side of the head associated with other disabling symptoms, such as nausea, increased sensitivity to light, sound and smell and mood changes. Various clinical factors, including the excessive use of migraine medication, inadequate acute treatment and stressful events, can contribute to the worsening of the condition, which may evolve to chronic migraine, that is, a headache present on >15 days/month for at least 3 months. Chronic migraine is frequently associated with various comorbidities, including anxiety and mood disorders, particularly depression, which complicate the prognosis, response to treatment and overall clinical outcomes. Emerging research indicates a connection between alterations in the composition of the gut microbiota and mental health conditions, particularly anxiety and depression, which are considered disorders of the gut-brain axis. This underscores the potential of modulating the gut microbiota as a new avenue for managing these conditions. In this context, it is interesting to investigate whether migraine, particularly in its chronic form, exhibits a dysbiosis profile similar to that observed in individuals with anxiety and depression. This could pave the way for interventions aimed at modulating the gut microbiota for treating difficult-to-manage migraines.
Asunto(s)
Microbioma Gastrointestinal , Trastornos Migrañosos , Humanos , Trastornos Migrañosos/microbiología , Trastornos Migrañosos/terapia , Trastornos Migrañosos/psicología , Eje Cerebro-Intestino , Ansiedad/microbiología , Depresión/microbiología , Disbiosis/microbiología , AnimalesRESUMEN
Psychological stress during adolescence may cause enduring cognitive deficits and anxiety in both humans and animals, accompanied by rearrangement of numerous brain structures and functions. A healthy diet is essential for proper brain development and maintenance of optimal cognitive functions during adulthood. Furthermore, nutritional components profoundly affect the intestinal community of microbes that may affect gut-brain communication. We adopted a relatively mild stress protocol, social instability stress, which when repeatedly administered to juvenile rats modifies cognitive behaviors and plasticity markers in the brain. We then tested the preventive effect of a prolonged diet enriched with the ω-3 polyunsaturated fatty acids eicosapentaenoic acid, docosahexaenoic acid, and docosapentaenoic acid and vitamin A. Our findings highlight the beneficial effects of this enriched diet on cognitive memory impairment induced by social instability stress, as stressed rats fed the enriched diet exhibited performance undistinguishable from that of nonstressed rats on both emotional and reference memory tests. Furthermore, in stressed rats, the decline in brain-derived neurotrophic factor expression in the hippocampus and shifts in the microbiota composition were normalized by the enriched diet. The detrimental behavioral and neurochemical effects of adolescent stress, as well as the protective effect of the enriched diet, were maintained throughout adulthood, long after the exposure to the stressful environment was terminated. Taken together, our results strongly suggest a beneficial role of nutritional components in ameliorating stress-related behaviors and associated neurochemical and microbiota changes, opening possible new venues in the field of nutritional neuropsychopharmacology.
Asunto(s)
Cognición/efectos de los fármacos , Dieta , Ácidos Grasos Omega-3/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Hipocampo/fisiopatología , Estrés Psicológico , Animales , Ansiedad/microbiología , Ansiedad/fisiopatología , Ansiedad/prevención & control , Conducta Animal/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Estrés Psicológico/microbiología , Estrés Psicológico/fisiopatología , Estrés Psicológico/prevención & controlRESUMEN
BACKGROUND: Anxiety and depression are complications in Irritable bowel syndrome (IBS) patients. In this study, we recruited 18 IBS patients with mild-modest anxiety and depression behaviors, and after the screening, we defined the FMT treatment group (n = 9) and the control group (n = 9). The IBS symptom severity scale (IBS-SSS), Hamilton Anxiety Rating Scale (HAM-A), Hamilton Depression Rating Scale (HAM-D), Irritable Bowel Syndrome Quality of Life (IBS-QOL) and Bristol stool scale (BSS) were evaluated one week before FMT (baseline), one-week-, one-month-, two-month-, and three-month-following FMT. Meanwhile, we determined the SCFAs in the patient's feces and serum and continued the metagenomic analysis of the microorganisms in the patient's feces. RESULTS: The results showed that the patient's anxiety and depression behavior gradually improved with FMT treatment. Moreover, the illness and quality of life had also been relieved significantly. The content of isovaleric acid and valeric acid was significantly reduced in the FMT group compared to the Col group. Metagenomic analysis showed that FMT treatment decreased the abundance of Faecalibacterium, Eubacterium and Escherichia. From KEGG functional analysis, we confirmed that the top five abundant pathways were "bacterial chemotaxis, "flagellar assembly", "glycine, serine and threonine metabolism", "apoptosis", and "bacterial invasion of epithelial cells". CONCLUSIONS: FMT treatment can effectively alleviate the anxiety and depression behaviors of IBS-D patients and reduce the IBS-SSS score, indicating that FMT can improve patients' symptoms. The high throughput sequencing results show that Bifidobacterium and Escherichia play the most critical role in the formation and recovery of IBS-D patients. The GC/MS data indicated that faeces isovaleric acid and valeric acid might be more suitable as a metabolic indicator of IBS-D remission. Trial registration ChiCTR, ChiCTR1900024924, Registered 3 August 2019, https://www.chictr.org.cn/showproj.aspx?proj=41676 .
Asunto(s)
Ansiedad/microbiología , Ansiedad/terapia , Depresión/microbiología , Depresión/terapia , Trasplante de Microbiota Fecal , Síndrome del Colon Irritable/microbiología , Metagenoma , Adulto , Anciano , Diarrea/microbiología , Diarrea/terapia , Escherichia/clasificación , Eubacterium/clasificación , Faecalibacterium/clasificación , Heces/microbiología , Femenino , Microbioma Gastrointestinal , Hemiterpenos/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/terapia , Masculino , Persona de Mediana Edad , Ácidos Pentanoicos/metabolismo , Calidad de VidaRESUMEN
PURPOSE: The microbiome-gut-brain (MGB) axis provides a dynamic model to understand associations between the gut microbiota and psychoneurological comorbidities. The role of the MGB axis in cancer treatment-related psychoneurological symptoms (PNS) remains unknown. The purpose of this study was to conduct a systematic review of the existing literature to identify the influence of the gut microbiota on cancer and cancer treatment-related PNS and toxicities mediated by the MGB axis. METHODS: We searched the databases of PubMed, Embase, and Web of Science from their earliest records to October 2019. All studies identified in the database searches were screened by title and abstract, followed by a review of the full texts. The Johns Hopkins Nursing Evidence-Based Practice Model was adopted to assess the evidence levels and qualities; the Joanna Briggs Institute critical appraisal tools were used to assess the methodological quality and the possibility of bias for each included study. All the study findings were combined, synthesized, and presented through narrative format. RESULTS: Six studies were included in this systematic review. These studies primarily focused on cancer survivorship while receiving chemotherapy, and they were conducted between 2016 and 2019. The gut microbiome was assessed via fecal samples, which were analyzed using 16S rRNA sequencing approaches. With small-scale studies, the gut microbiota was associated with cancer treatment-related PNS, including fatigue, anxiety, depression, sleep disturbance, cognitive impairment, and chemotherapy-induced peripheral neuropathy. A higher relative abundance of Bacteroides was associated with a higher level of fear of cancer recurrence but a higher relative abundance of Lachnospiraceae.g and Ruminococcus was associated with a lower level in fear of cancer recurrence. Changes in fatigue interference were associated with the frequency of genera Faecalibacterium and Prevotella, and changes in anxiety were associated with the frequency of genera Coprococcus and Bacteroides. CONCLUSIONS: The gut microbiota showed significant associations with cancer treatment-related PNS. Recent work regarding the MGB axis in cancer psychoneurological toxicities focused primarily on individual toxicity and symptoms in cancer survivors with chemotherapy exposure. Associations between the gut microbiota and PNS should be further studied in cancer populations across different ages, cancer types, and treatment modalities.
Asunto(s)
Encéfalo/microbiología , Microbioma Gastrointestinal/fisiología , Neoplasias/microbiología , Neoplasias/psicología , Animales , Ansiedad/etiología , Ansiedad/microbiología , Supervivientes de Cáncer/psicología , Heces/microbiología , Humanos , Neoplasias/patología , Neoplasias Primarias SecundariasRESUMEN
Objective: Altering the gut microflora may produce health benefits in individuals suffering from mood disorders. The purpose of this review was to evaluate the efficacy of probiotics, prebiotics, or synbiotics as a potential treatment for symptoms of depression, anxiety, and stress (as psychobiotics).Methods: Google Scholar, PubMed, PsychINFO, and Web of Science were utilized to identify and evaluate studies through October 31, 2019. Studies were included if subjects were evaluated for altered mood or stress levels at start of the study and consumed probiotics, prebiotics, and/or synbiotics for intervention.Results: Search results yielded 142 articles, while only 12 studies met all inclusion criteria. Nine of the 12 studies identified evaluated the efficacy of various probiotic strains, while only two evaluated synbiotics and one evaluated prebiotics. Six out of 12 studies found probiotics to reduce depression, while two studies found probiotics to reduce anxiety.Discussion: Translational research in this field is limited and further investigation of the efficacy of psychobiotics in mood disorders is warranted.
Asunto(s)
Ansiedad/terapia , Depresión/terapia , Microbioma Gastrointestinal/fisiología , Prebióticos/administración & dosificación , Probióticos/uso terapéutico , Simbióticos/administración & dosificación , Ansiedad/microbiología , Depresión/microbiología , Humanos , Estrés Psicológico/microbiología , Estrés Psicológico/terapiaRESUMEN
BACKGROUND: The aim of this study was to investigate the effects of probiotic and synbiotic supplementation on the depression and anxiety symptoms and serum brain-derived neurotrophic factor (BDNF) level. METHODS: Seventy-five HD patients were randomly assigned to receive the synbiotic (15â g of prebiotics, 5â g of probiotic containing Lactobacillus acidophilus T16, Bifidobacterium bifidum BIA-6, Bifidobacterium lactis BIA-7, and Bifidobacterium longum BIA-8 (2.7 × 107â CFU/g each)) or probiotics (5â g probiotics as in synbiotic group with 15â g of maltodextrin as placebo) or placebo (20â g of maltodextrin) for 12 weeks. Serum BDNF was measured by ELISA kit. Hospital Anxiety and Depression Scale (HADS) was used to assess symptoms of depression (HADS-DEP) and anxiety (HADS-ANX). RESULTS: From baseline to 12 weeks, synbiotic supplementation resulted in a significant decrease in HADS-DEP score in a subgroup of patients with depressive symptom (HADS-DEP ≥ 8) compared to the placebo and probiotic supplementation (p = .001, p = .002, respectively) and in all patients compared to the placebo (p = .004). There was no significant difference among the groups in terms of HADS-ANX scores. However, the HADS-ANX scores decreased significantly in the synbiotic group compared to the baseline in all patients (p = .047) and also patients with depressive symptom (p = .03). In addition, in a subgroup of HD patients with depressive symptom, the serum BDNF increased significantly in the synbiotic group when compared to the placebo (p < .001) and probiotic group (p = .011). CONCLUSION: Overall, 12 weeks of synbiotic supplementation resulted in greater improvement in depression symptoms and serum BDNF level compared to the probiotic supplementation in HD patients especially in the subgroup of patients with depression symptoms.
Asunto(s)
Ansiedad/sangre , Factor Neurotrófico Derivado del Encéfalo/sangre , Depresión/sangre , Suplementos Dietéticos , Enfermedades Renales/complicaciones , Probióticos/administración & dosificación , Diálisis Renal , Simbióticos/administración & dosificación , Adulto , Ansiedad/microbiología , Depresión/microbiología , Método Doble Ciego , Femenino , Humanos , Enfermedades Renales/sangre , Enfermedades Renales/psicología , Masculino , Persona de Mediana Edad , Resultado del TratamientoRESUMEN
ABSTRACT: Burgeoning body of evidence from neuroscience is pouring in highlighting a potential association between gut microbiota with the pathophysiology of depression and anxiety. Manipulation of gut microbiota may be then useful to decode this role and to provide novel therapeutics for major depressive disorder (MDD), developing microbiota-related biomarkers to stratify patients at risk and to delineate more homogeneous biotypes of MDD.
Asunto(s)
Trastorno Depresivo Mayor/microbiología , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped , Animales , Ansiedad/microbiología , Biomarcadores , Depresión/microbiología , Trastorno Depresivo Mayor/diagnóstico , Trastorno Depresivo Mayor/dietoterapia , Humanos , Ratones , Prebióticos , Probióticos/uso terapéutico , RatasRESUMEN
Anxiety and eating disorders produce a physiological imbalance that triggers alterations in the abundance and composition of gut microbiota. Moreover, the gut-brain axis can be altered by several factors such as diet, lifestyle, infections, and antibiotic treatment. Diet alterations generate gut dysbiosis, which affects immune system responses, inflammation mechanisms, the intestinal permeability, as well as the production of short chain fatty acids and neurotransmitters by gut microbiota, which are essential to the correct function of neurological processes. Recent studies indicated that patients with generalized anxiety or eating disorders (anorexia nervosa, bulimia nervosa, and binge-eating disorders) show a specific profile of gut microbiota, and this imbalance can be partially restored after a single or multi-strain probiotic supplementation. Following the PRISMA methodology, the current review addresses the main microbial signatures observed in patients with generalized anxiety and/or eating disorders as well as the importance of probiotics as a preventive or a therapeutic tool in these pathologies.
Asunto(s)
Trastornos de Ansiedad/microbiología , Ansiedad/microbiología , Trastornos de Alimentación y de la Ingestión de Alimentos/microbiología , Probióticos/uso terapéutico , Animales , Ansiedad/terapia , Trastornos de Ansiedad/terapia , Trastornos de Alimentación y de la Ingestión de Alimentos/terapia , Microbioma Gastrointestinal , HumanosRESUMEN
BACKGROUND: Psychological co-morbidities in irritable bowel syndrome (IBS) have been widely recognized, whereas less is known regarding the role of gut microbial and host metabolic changes in clinical and psychological symptoms in IBS. RESULTS: A total of 70 diarrhoea-predominant IBS (IBS-D) patients and 46 healthy controls were enrolled in this study. Stool and urine samples were collected from both groups for 16S rRNA gene sequencing and metabolomic analysis. The results showed that fecal microbiota in IBS-D featured depleted Faecalibacterium (adjusted P = 0.034), Eubacterium rectale group (adjusted P = 0.048), Subdoligranulum (adjusted P = 0.041) and increased Prevotella (adjusted P = 0.041). O-ureido-L-serine, 3,4-dihydroxybenzenesulfonic acid and (R)-2-Hydroxyglutarate demonstrated lower urinary concentrations in IBS-D patients. We further built correlation matrices between gut microbe abundance, differentiated metabolite quantities and clinical parameters. Dialister manifested negative association with IBS severity (r = - 0.285, P = 0.017), anxiety (r = - 0.347, P = 0.003) and depression level (r = - 0.308, P = 0.010). Roseburia was negatively associated with IBS severity (r = - 0.298, P = 0.012). Twenty metabolites correlated with anxiety or depression levels, including 3,4-dihydroxymandelaldehyde with SAS (r = - 0.383, P = 0.001), 1-methylxanthine with SDS (r = - 0.347, P = 0.004) and 1D-chiro-inositol with SAS (r = - 0.336, P = 0.005). In analysis of microbe-metabolite relationship, 3,4-dihydroxymandelaldehyde and 1-methylxanthine were negatively correlated with relative abundance of Clostridiumsensu stricto. CONCLUSIONS: Our findings demonstrated altered microbial and metabolomic profiles associated with clinically and psychological symptoms in IBS-D patients, which may provide insights for further investigations.
Asunto(s)
Ansiedad/microbiología , Bacterias/clasificación , Depresión/microbiología , Diarrea/psicología , Síndrome del Colon Irritable/psicología , Metabolómica/métodos , Análisis de Secuencia de ADN/métodos , Adulto , Ansiedad/metabolismo , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , Comorbilidad , ADN Bacteriano/genética , ADN Ribosómico/genética , Depresión/metabolismo , Diarrea/metabolismo , Diarrea/microbiología , Heces/microbiología , Femenino , Glutaratos/orina , Homoserina/análogos & derivados , Homoserina/orina , Humanos , Síndrome del Colon Irritable/metabolismo , Síndrome del Colon Irritable/microbiología , Masculino , Persona de Mediana Edad , Filogenia , ARN Ribosómico 16S/genética , Orina/química , Orina/microbiología , Xantinas/orinaRESUMEN
Objective: The intestinal microbiota is acknowledged to be essential in brain development and behaviour. Their composition can be modulated by prebiotics such as short-chain galacto-oligosaccharides (scGOS) and long-chain fructo-oligosaccharide (lcFOS). Several studies reported potential health benefit of prebiotics on behaviour. As the prebiotic mixture of scGOS and lcFOS is included in infant formula, we investigated the effects of dietary supplementation with this specific mixture from the day of birth onwards on behaviour and intestinal microbiota development in mice. Method: Healthy male BALB/cByJ mice received, from day of birth, a dietary supplement with or without 3% scGOS:lcFOS (9:1). Behavioural tests were performed pre-weaning, in adolescence, early adulthood and adulthood. We assessed faecal microbiota compositions over time, caecal short-chain fatty acids as well as brain mRNA expression of Htr1a, Htr1b and Tph2 and monoamine levels. Results: Compared to control fed mice, scGOS:lcFOS fed mice showed reduced anxiety-like and repetitive behaviour over time and improved social behaviour in adulthood. The serotonergic system in the prefrontal cortex (PFC) and somatosensory cortex (SSC) was affected by the scGOS:lcFOS. In the PFC, mRNA expression of brain-derived neurotrophic factor (Bdnf) was enhanced in scGOS:lcFOS fed mice. Although the bacterial diversity of the intestinal microbiota was unaffected by the scGOS:lcFOS diet, microbiota composition differed between the scGOS:lcFOS and the control fed mice over time. Moreover, an increased saccharolytic and decreased proteolytic fermentation activity were observed in caecum content. Discussion: Supplementing the diet with scGOS:lcFOS from the day of birth is associated with reduced anxiety-like and improved social behaviour during the developmental period and later in life, and modulates the composition and activity of the intestinal microbiota in healthy male BALB/c mice. These data provide further evidence of the potential impact of scGOS:lcFOS on behaviour at several developmental stages throughout life and strengthen the insights in the interplay between the developing intestine and brain.
Asunto(s)
Ansiedad/microbiología , Microbioma Gastrointestinal , Oligosacáridos/administración & dosificación , Prebióticos/administración & dosificación , Conducta Social , Animales , Ansiedad/prevención & control , Conducta Animal , Monoaminas Biogénicas/metabolismo , Encéfalo/metabolismo , Masculino , Ratones Endogámicos BALB C , Vocalización AnimalRESUMEN
Objective: Pre-clinical evidence suggests that the gastrointestinal microbiota contributes to mood and behavior disorders. Among humans, diet quality and patterns, which also impact the gastrointestinal microbiota, have been linked to depression, anxiety, and stress. This review summarizes findings from clinical studies using dietary intervention to improve depression, anxiety, or stress and the role the gastrointestinal microbiota may have in these disorders.Methods: A literature search was conducted using the keywords microbiome, microbiota, depression, anxiety, stress, diet, dietary pattern, diet quality, fiber, prebiotics, probiotics, and mood.Results: Mood was improved by enhancing diet quality. Fructooligosaccharide and galactooligosaccharide improved anxiety and depression in participants consuming ≥ 5â g/day. Additionally, bifidobacteria were enriched in subjects consuming ≥ 5â g/day. Probiotic consumption improved psychological or biological measures of depression, anxiety, or stress in individuals predisposed to a mood disorder. Probiotics suppressed biological markers of stress in healthy individuals in a strain-dependent manner.Discussion: High-quality diets, prebiotics, and probiotics may beneficially affect mood. Habitual diets rich in dietary fiber and omega-3-polyunsaturated fatty acids may be linked to reduced risk of developing symptoms of depression, anxiety, and stress; however, additional studies are necessary. Certain probiotics may enhance mood, but their influence on the gastrointestinal microbiota requires further investigation.
Asunto(s)
Ansiedad/epidemiología , Depresión/epidemiología , Dieta , Microbioma Gastrointestinal/fisiología , Estrés Psicológico/epidemiología , Afecto/fisiología , Ansiedad/microbiología , Ansiedad/prevención & control , Depresión/microbiología , Depresión/prevención & control , Dieta Saludable , Dieta Mediterránea , Fibras de la Dieta/administración & dosificación , Ácidos Grasos Omega-3/administración & dosificación , Humanos , Prebióticos/administración & dosificación , Probióticos/administración & dosificación , Estrés Psicológico/microbiología , Estrés Psicológico/prevención & controlRESUMEN
Tuberculosis (TB) is a chronic infectious disease in which prolonged, non-resolutive inflammation of the lung may lead to metabolic and neuroendocrine dysfunction. Previous studies have reported that individuals coursing pulmonary TB experience cognitive or behavioural changes; however, the pathogenic substrate of such manifestations have remained unknown. Here, using a mouse model of progressive pulmonary TB, we report that, even in the absence of brain infection, TB is associated with marked increased synthesis of both inflammatory and anti-inflammatory cytokines in discrete brain areas such as the hypothalamus, the hippocampal formation and cerebellum accompanied by substantial changes in the synthesis of neurotransmitters. Moreover, histopathological findings of neurodegeneration and neuronal death were found as infection progressed with activation of p38, JNK and reduction in the BDNF levels. Finally, we perform behavioural analysis in infected mice throughout the infection, and our data show that the cytokine and neurochemical changes were associated with a marked onset of cognitive impairment as well as depressive- and anxiety-like behaviour. Altogether, our results suggest that besides pulmonary damage, TB is accompanied by an extensive neuroinflammatory and neurodegenerative state which explains some of the behavioural abnormalities found in TB patients.
Asunto(s)
Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Mycobacterium tuberculosis/metabolismo , Neuronas/patología , Tuberculosis Pulmonar/metabolismo , Animales , Ansiedad/metabolismo , Ansiedad/microbiología , Síntomas Conductuales/microbiología , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/citología , Encéfalo/enzimología , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión , Disfunción Cognitiva/microbiología , Depresión/metabolismo , Depresión/microbiología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Hipocampo/citología , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Masculino , Ratones Endogámicos BALB C , Mycobacterium tuberculosis/patogenicidad , Neuronas/citología , Neurotransmisores/metabolismo , Tuberculosis Pulmonar/enzimología , Tuberculosis Pulmonar/patología , Tuberculosis Pulmonar/psicología , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismoRESUMEN
Recent studies have suggested the neuroprotective effects of Clostridium butyricum on mood disorders. However, the potential role of Clostridium butyricum in modulating the gut-brain-axis remains unknown. Here, we applied the commercial Clostridium butyricum Miyairi 588 (CBM588) strain to assess psychological behavioural alterations in mice exposed to chronic social defeat stress (CSDS). We found that preventive treatment with CBM588 for 28 days ameliorated depressive-like behaviours in CSDS mice. We showed that CSDS led to increases in cytokines (IL-1ß, IL-6, and TNF-α), intestinal dysfunction and hippocampal microglial activation, while CBM588 partially relieved these alterations. By applying 16S sequencing, we found that Firmicutes was more abundant in the faeces of CBM588/CSDS mice than in the faeces of placebo/CSDS mice, and depression-like behaviours in the mice were correlated with certain strains (including Clostridium leptum, Blautia coccoides, Family_XIII_UCG-001, Candidatus Arthromitus sp-SFB-mouse-Japan and Streptococcus hyointestinalis) at the species level. Our results illustrated the preventive effect of CBM588 against stress, suggesting the beneficial role of CBM588 in regulating neuroinflammation via the gut-brain-axis. This study provides novel strategies for clinical and scientific investigations of depressive disorders.