Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 187
Filtrar
1.
PLoS One ; 19(6): e0304787, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38837966

RESUMEN

OBJECTIVE: The objective of this scoping review is to review the research evidence regarding the impact of perinatal maternal stress on the maternal and infant gut and human milk microbiomes. INTRODUCTION: Perinatal stress which refers to psychological stress experienced by individuals during pregnancy and the postpartum period is emerging as a public health concern. Early exposure of infants to perinatal maternal stress can potentially lead to metabolic, immune, and neurobehavioral disorders that extend into adulthood. The role of the gut and human milk microbiome in the microbiome-gut-brain axis as a mechanism of stress transfer has been previously reported. A transfer of colonised aberrant microbiota from mother to infant is proposed to predispose the infant to a pro- inflammatory microbiome with dysregulated metabolic process thereby initiating early risk of chronic diseases. The interplay of perinatal maternal stress and its relationship to the maternal and infant gut and human milk microbiome requires further systematic examination in the literature. INCLUSION CRITERIA: This scoping review is an exploratory mapping review which will focus on the population of mothers and infants with the exploration of the key concepts of maternal stress and its impact on the maternal and infant gut and human milk microbiome in the context of the perinatal period. It will focus on the pregnancy and the post-natal period up to 6 months with infants who are exclusively breastfed. METHODS: This study will be guided by the Joanna Briggs Institute's (JBI) methodology for scoping reviews along with use of the Prisma Scr reporting guideline. A comprehensive search will be conducted using the following databases, CINAHL Complete; MEDLINE; PsycINFO, Web of Science and Scopus. A search strategy with pre-defined inclusion and exclusion criteria will be used to retrieve peer reviewed data published in English from 2014 to present. Screening will involve a three-step process with screening tool checklists. Results will be presented in tabular and narrative summaries, covering thematic concepts and their relationships. This protocol is registered with Open Science Framework DOI 10.17605/OSF.IO/5SRMV.


Asunto(s)
Microbioma Gastrointestinal , Leche Humana , Estrés Psicológico , Humanos , Leche Humana/microbiología , Femenino , Embarazo , Estrés Psicológico/microbiología , Lactante , Recién Nacido , Lactancia Materna , Madres/psicología
2.
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
3.
Pharmacol Res ; 204: 107214, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38763328

RESUMEN

Studies have shown that the microbiota-gut-brain axis is highly correlated with the pathogenesis of depression in humans. However, whether independent oral microbiome that do not depend on gut microbes could affect the progression of depression in human beings remains unclear, neither does the presence and underlying mechanisms of the microbiota-oral-brain axis in the development of the condition. Hence this study that encompasses clinical and animal experiments aims at investigating the correlation between oral microbiota and the onset of depression via mediating the microbiota-oral-brain axis. We compared the oral microbial compositions and metabolomes of 87 patients with depressive symptoms versus 70 healthy controls. We found that the oral microbial and metabolic signatures were significantly different between the two groups. Significantly, germ-free (GF) mice transplanted with saliva from mice exposing to chronic restraint stress (CRS) displayed depression-like behavior and oral microbial dysbiosis. This was characterized by a significant differential abundance of bacterial species, including the enrichment of Pseudomonas, Pasteurellaceae, and Muribacter, as well as the depletion of Streptococcus. Metabolomic analysis showed the alternation of metabolites in the plasma of CRS-exposed GF mice, especially Eicosapentaenoic Acid. Furthermore, oral and gut barrier dysfunction caused by CRS-induced oral microbiota dysbiosis may be associated with increased blood-brain barrier permeability. Pseudomonas aeruginosa supplementation exacerbated depression-like behavior, while Eicosapentaenoic Acid treatment conferred protection against depression-like states in mice. These results suggest that oral microbiome and metabolic function dysbiosis may be relevant to the pathogenesis and pathophysiology of depression. The proposed microbiota-oral-brain axis provides a new way and targets for us to study the pathogenesis of depression.


Asunto(s)
Depresión , Disbiosis , Estrés Psicológico , Animales , Disbiosis/metabolismo , Depresión/metabolismo , Depresión/microbiología , Depresión/psicología , Depresión/etiología , Masculino , Humanos , Estrés Psicológico/metabolismo , Estrés Psicológico/microbiología , Estrés Psicológico/psicología , Femenino , Adulto , Ratones , Restricción Física/psicología , Ratones Endogámicos C57BL , Microbioma Gastrointestinal , Eje Cerebro-Intestino , Boca/microbiología , Persona de Mediana Edad , Saliva/metabolismo , Saliva/microbiología , Conducta Animal , Barrera Hematoencefálica/metabolismo
4.
Commun Biol ; 7(1): 670, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822061

RESUMEN

Stress in early life can affect the progeny and increase the risk to develop psychiatric and cardiometabolic diseases across generations. The cross-generational effects of early life stress have been modeled in mice and demonstrated to be associated with epigenetic factors in the germline. While stress is known to affect gut microbial features, whether its effects can persist across life and be passed to the progeny is not well defined. Here we show that early postnatal stress in mice shifts the fecal microbial composition (binary Jaccard index) throughout life, including abundance of eight amplicon sequencing variants (ASVs). Further effects on fecal microbial composition, structure (weighted Jaccard index), and abundance of 16 ASVs are detected in the progeny across two generations. These effects are not accompanied by changes in bacterial metabolites in any generation. These results suggest that changes in the fecal microbial community induced by early life traumatic stress can be perpetuated from exposed parent to the offspring.


Asunto(s)
Heces , Microbioma Gastrointestinal , Estrés Psicológico , Animales , Heces/microbiología , Ratones , Estrés Psicológico/microbiología , Femenino , Masculino , Ratones Endogámicos C57BL , Bacterias/genética , Bacterias/clasificación
6.
Cell Metab ; 36(3): 466-483.e7, 2024 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-38266651

RESUMEN

The brain and gut are intricately connected and respond to various stimuli. Stress-induced brain-gut communication is implicated in the pathogenesis and relapse of gut disorders. The mechanism that relays psychological stress to the intestinal epithelium, resulting in maladaptation, remains poorly understood. Here, we describe a stress-responsive brain-to-gut metabolic axis that impairs intestinal stem cell (ISC) lineage commitment. Psychological stress-triggered sympathetic output enriches gut commensal Lactobacillus murinus, increasing the production of indole-3-acetate (IAA), which contributes to a transferrable loss of intestinal secretory cells. Bacterial IAA disrupts ISC mitochondrial bioenergetics and thereby prevents secretory lineage commitment in a cell-intrinsic manner. Oral α-ketoglutarate supplementation bolsters ISC differentiation and confers resilience to stress-triggered intestinal epithelial injury. We confirm that fecal IAA is higher in patients with mental distress and is correlated with gut dysfunction. These findings uncover a microbe-mediated brain-gut pathway that could be therapeutically targeted for stress-driven gut-brain comorbidities.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Linaje de la Célula , Estrés Psicológico/microbiología , Acetatos , Indoles/farmacología
7.
Clin Transl Oncol ; 26(6): 1407-1418, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38194019

RESUMEN

INTRODUCTION: Breast cancer (BC) is the most prevalent type of cancer and has the highest mortality among women worldwide. BC patients have a high risk of depression, which has been recognized as an independent factor in the progression of BC. However, the potential mechanism has not been clearly demonstrated. METHODS: To explore the correlation and mechanism between depression and BC progression, we induced depression and tumor in BC mouse models. Depression was induced via chronic unpredictable mild stress (CUMS) and chronic restraint stress (CRS). Amino acid (AA) neurotransmitter-targeted metabonomics and gut microbiota 16S rDNA gene sequencing were employed in the mouse model after evaluation with behavioral tests and pathological analysis. RESULTS: The tumors in cancer-depression (CD) mice grew faster than those in cancer (CA) mice, and lung metastasis was observed in CD mice. Metabonomics revealed that the neurotransmitters and plasma AAs in CD mice were dysregulated, namely the tyrosine and tryptophan pathways and monoamine neurotransmitters in the brain. Gut microbiota analysis displayed an increased ratio of Firmicutes/Bacteroides. In detail, the abundance of f_Lachnospiraceae and s_Lachnospiraceae increased, whereas the abundance of o_Bacteroidales and s_Bacteroides_caecimuris decreased. Moreover, the gut microbiota was more closely associated with AA neurotransmitters than with plasma AA. CONCLUSION: Depression promoted the progression of BC by modulating the abundance of s_Lachnospiraceae and s_Bacteroides_caecimuris, which affected the metabolism of monoamine neurotransmitters in the brain and AA in the blood.


Asunto(s)
Aminoácidos , Neoplasias de la Mama , Depresión , Progresión de la Enfermedad , Microbioma Gastrointestinal , Neurotransmisores , Animales , Microbioma Gastrointestinal/fisiología , Femenino , Ratones , Neurotransmisores/metabolismo , Aminoácidos/metabolismo , Depresión/metabolismo , Depresión/microbiología , Neoplasias de la Mama/patología , Neoplasias de la Mama/microbiología , Neoplasias de la Mama/metabolismo , Metabolómica , Modelos Animales de Enfermedad , Estrés Psicológico/microbiología , Estrés Psicológico/metabolismo , Estrés Psicológico/complicaciones
8.
Dev Psychobiol ; 65(7): e22424, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37860905

RESUMEN

Prenatal maternal stress (PNMS)-characterized by exposure to stress, anxiety, depression, or intimate partner violence-has been linked to biological alterations in infants, including disruptions to their intestinal microbiota, which have long-term implications for children's developmental outcomes. Significant research has been done examining the effects of PNMS on the microbiome in animals, but less is known about these effects in human research. The current systematic review aimed to synthesize current findings on the association between PNMS and mother and infant microbiomes. Medline, Embase, PsycInfo, Web of Science, and Eric databases were searched through to February 2022. A total of eight studies (n = 2219 infants, 2202 mothers) were included in the qualitative synthesis. Findings provided promising evidence of the role that PNMS plays in altering the microbial composition, diversity, and gut immunity in mothers and infants. Notably, majority of included studies found that higher PNMS was linked to increases in genera from the phylum Proteobacteria. The factors influencing these effects are explored including nutrition, birth mode, and parenting behaviors. Potential interventions to mitigate the adverse effects of PNMS are discussed, along with recommendations for future studies with longitudinal designs to better understand the appropriate type and timing of interventions needed to promote "healthy" maternal and infant microbial functioning.


Asunto(s)
Microbioma Gastrointestinal , Madres , Femenino , Niño , Embarazo , Animales , Humanos , Lactante , Estrés Psicológico/microbiología , Ansiedad , Trastornos de Ansiedad
9.
Front Cell Infect Microbiol ; 13: 1121445, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36909723

RESUMEN

Backgrounds: Gut microbiota plays a critical role in the onset and development of depression, but the underlying molecular mechanisms are unclear. This study was conducted to explore the relationships between gut microbiota and host's metabolism in depression. Methods: Chronic social defeat stress (CSDS) model of depression was established using C57BL/6 male mice. Fecal samples were collected from CSDS group and control group to measure gut microbiota and microbial metabolites. Meanwhile, tryptophan metabolism-related metabolites in hippocampus were also analyzed. Results: CSDS successfully induced depressive-like behaviors in CSDS group. The 24 differential bacterial taxa between the two groups were identified, and 14 (60.87%) differential bacterial taxa belonged to phylum Firmicutes. Functional analysis showed that tryptophan metabolism was significantly affected in CSDS mice. Meanwhile, 120 differential microbial metabolites were identified, and two key tryptophan metabolism-related metabolites (tryptophan and 5-hydroxytryptophan (5-HTP)) were significantly decreased in feces of CSDS mice. The correlation analysis found the significant relationships between tryptophan and differential bacterial taxa under Firmicutes, especially genus Lactobacillus (r=0.801, p=0.0002). In addition, the significantly decreased 5-hydroxytryptamine (5-HT) in hippocampus of depressed mice was also observed. Conclusions: Our results showed that tryptophan metabolism might have an important role in the crosstalk between gut microbioa and brain in depression, and phylum Firmicutes, especially genus Lactobacillus, might be involved in the onset of depression through regulating tryptophan metabolism.


Asunto(s)
Depresión , Microbioma Gastrointestinal , Ratones , Masculino , Animales , Depresión/metabolismo , Depresión/microbiología , Triptófano , Derrota Social , Ratones Endogámicos C57BL , Encéfalo , Bacterias , Estrés Psicológico/microbiología
10.
Food Res Int ; 157: 111289, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35761597

RESUMEN

Tryptophan, an essential amino acid, has been reported that it has the potential to regulate depression-like behavior. Meanwhile, Chronic stress-induced depression also has a close relationship with gut microbiota structure and composition. In the current research, we demonstrated that a tryptophan-rich diet (0.6% tryptophan w/w) significantly attenuated depression- and anxiety-like behaviors in a chronic unpredictable mild stress (CUMS)-treated mouse model. Tryptophan supplementation improved neuroinflammation, increased expression of BDNF, and improved mitochondrial energy metabolism in the brain of CUMS-treated mice. Besides, CUMS also enhanced the kynurenine pathway, but repressed the serotonin pathway and indole pathway of tryptophan metabolism, leading to a decrease in 5-HT and indole in serum, whereas tryptophan supplementation might shift the tryptophan metabolism more toward the serotonin pathway in CUMS-treated mice. The gut microbiome was restructured by increasing the relative abundance of Lachnospiracea, Clostridium, Lactobacillus, Bifidobacterium in tryptophan-treated depressive mice. Moreover, tryptophan administration inhibited stress-induced gut barrier damage and decreased inflammatory responses in the colon. Together, our study purports the gut-brain axis as a mechanism for the potential of tryptophan to improve depression and anxiety-related behavior.


Asunto(s)
Depresión , Triptófano , Animales , Ansiedad , Conducta Animal , Eje Cerebro-Intestino , Depresión/metabolismo , Dieta , Ratones , Serotonina , Estrés Psicológico/metabolismo , Estrés Psicológico/microbiología
11.
Psychoneuroendocrinology ; 136: 105620, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34896741

RESUMEN

Antibiotics exposure leads to gut microbiota dysbiosis, which increases the risk of anxiety and depression. However, the impact of ciprofloxacin and metronidazole exposure on chronic unpredictable mild stress-induced anxiety-like and depression-like behavior and underlying regulatory mechanism have not been well established. Here, chronic unpredictable mild stress model was established in adult male Sprague-Dawley rats. 16 S rRNA gene sequencing was used to decipher the gut microbiota. Enzyme-linked immunosorbent assay (ELIZA) was used to measure circulating cytokines in blood, gut barrier permeability biomarkers in feces, blood-brain barrier permeability biomarkers in brain. We found that antibiotics exposure significantly reduced the body weight, weight gain and liver health in chronic unpredictable mild stress treated rats. Behavioral testing suggested that antibiotics exposure reduced anxiety-like and depression-like behavior of rat. Antibiotics exposure possessed lower bacterial richness and diversity than that in the chronic unpredictable mild stress treated group. Compared with CUMS or CUMS-e group, higher abundances of Bacteroides, Lactobacillus, Lachnospiraceae and Akkermansia, lower abundances of S24-7, Blautia, Ruminocaceae, Ruminococcus and Prevotella were found in the gut microbiota from antibiotics exposure group. In addition, short-term antibiotics exposure increased the level of 5-hydroxytryptamine (5-HT) in brain. A significant correlation between certain bacteria and behavior of rats was observed, such as Roseburia. Our study uncovers the role for antibiotics in regulating chronic unpredictable mild stress-induced anxiety-like and depression-like behavior and suggest that short-term antibiotics exposure may be could reverse chronic unpredictable mild stress-induced anxiety-like and depression-like behavior.


Asunto(s)
Antibacterianos , Depresión , Animales , Antibacterianos/farmacología , Ansiedad/tratamiento farmacológico , Conducta Animal , Depresión/tratamiento farmacológico , Depresión/microbiología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Serotonina , Estrés Psicológico/complicaciones , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/microbiología
12.
PLoS One ; 16(12): e0260813, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34890405

RESUMEN

Molecular-bacterial vaginosis (BV) is characterized by low levels of vaginal Lactobacillus species and is associated with higher risk of sexually transmitted infections (STI). Perceived psychosocial stress is associated with increased severity and persistence of infections, including STIs. American Indians have the highest rates of stress and high rates of STIs. The prevalence of molecular-BV among American Indian women is unknown. We sought to evaluate measures of psychosocial stress, such as historic loss (a multigenerational factor involving slavery, forced removal from one's land, legally ratified race-based segregation, and contemporary discrimination) and their association with the vaginal microbiota and specific metabolites associated with BV, in 70 Northwestern Plains American Indian women. Demographics, perceived psychosocial stressors, sexual practices, and known BV risk factors were assessed using a modified version of the American Indian Service Utilization, Psychiatric Epidemiology, Risk and Protective Factors Project survey. Self-collected mid-vaginal swabs were profiled for bacterial composition by 16S rRNA gene amplicon sequencing and metabolites quantified by targeted liquid-chromatography mass spectrometry. Sixty-six percent of the participants were classified as having molecular-BV, with the rest being either dominated by L. crispatus (10%) or L. iners (24%). High levels of lifetime trauma were associated with higher odds of having molecular-BV (adjusted Odds Ratio (aOR): 2.5, 95% Credible Interval (CrI): 1.1-5.3). Measures of psychosocial stress, including historic loss and historic loss associated symptoms, were significantly associated with lifestyle and behavioral practices. Higher scores of lifetime trauma were associated with increased concentrations of spermine (aFC: 3.3, 95% CrI: 1.2-9.2). Historic loss associated symptoms and biogenic amines were the major correlates of molecular-BV. Historical loss associated symptoms and lifetime trauma are potentially important underlying factors associated with BV.


Asunto(s)
Indio Americano o Nativo de Alaska/estadística & datos numéricos , Bacterias/clasificación , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN/métodos , Estrés Psicológico/epidemiología , Vaginosis Bacteriana/epidemiología , Adulto , Bacterias/genética , Bacterias/aislamiento & purificación , ADN Bacteriano/genética , ADN Ribosómico/genética , Femenino , Humanos , Microbiota , Persona de Mediana Edad , Filogenia , Prevalencia , Estrés Psicológico/microbiología , Estados Unidos/etnología , Vagina/microbiología , Vaginosis Bacteriana/microbiología , Adulto Joven
13.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34884735

RESUMEN

Stress disorders have dramatically increased in recent decades becoming the most prevalent psychiatric disorder in the United States and Europe. However, the diagnosis of stress disorders is currently based on symptom checklist and psychological questionnaires, thus making the identification of candidate biomarkers necessary to gain better insights into this pathology and its related metabolic alterations. Regarding the identification of potential biomarkers, omic profiling and metabolic footprint arise as promising approaches to recognize early biochemical changes in such disease and provide opportunities for the development of integrative candidate biomarkers. Here, we studied plasma and urine metabolites together with metagenomics in a 3 days Chronic Unpredictable Mild Stress (3d CUMS) animal approach that aims to focus on the early stress period of a well-established depression model. The multi-omics integration showed a profile composed by a signature of eight plasma metabolites, six urine metabolites and five microbes. Specifically, threonic acid, malic acid, alpha-ketoglutarate, succinic acid and cholesterol were proposed as key metabolites that could serve as key potential biomarkers in plasma metabolome of early stages of stress. Such findings targeted the threonic acid metabolism and the tricarboxylic acid (TCA) cycle as important pathways in early stress. Additionally, an increase in opportunistic microbes as virus of the Herpesvirales was observed in the microbiota as an effect of the primary stress stages. Our results provide an experimental biochemical characterization of the early stage of CUMS accompanied by a subsequent omic profiling and a metabolic footprinting that provide potential candidate biomarkers.


Asunto(s)
Metaboloma , Microbiota , Estrés Psicológico/metabolismo , Animales , Biomarcadores/sangre , Biomarcadores/orina , Masculino , Ratas Wistar , Estrés Psicológico/microbiología
14.
Gut Microbes ; 13(1): 2000275, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34856844

RESUMEN

An array of chronic inflammatory diseases, including metabolic diseases such as obesity and diabetes, are thought to be promoted by disturbance of the intestinal microbiota. Such diseases disproportionately impact low-income communities, which are frequently afflicted by chronic stress and increased density housing. Hence, we hypothesized that overcrowded housing might promote stress, microbiota dysbiosis, inflammation, and, consequently, metabolic diseases. We tested this hypothesis in a tractable murine model of social overcrowding (SOC), in which mice were housed at twice normal density. SOC moderately impacted behavior in some widely used assays (Open Field, Elevated Plus Maze and Light/Dark tests) and resulted in a stark increase in corticosterone levels. Such indices of stress were associated with mild chronic gut inflammation, hyperglycemia, elevations in colonic cytokines, and alterations in gut microbiota composition. All of these consequences of SOC were eliminated by broad spectrum antibiotics, while some (inflammation and hyperglycemia) were transmitted by microbiota transplantation from SOC mice to germfree mice housed at normal density. Altogether, these results suggest a central role for intestinal microbiota in driving stress, inflammation, and chronic diseases that are promoted by overcrowded housing.


Asunto(s)
Aglomeración/psicología , Microbioma Gastrointestinal , Estrés Psicológico/metabolismo , Estrés Psicológico/microbiología , Animales , Antibacterianos/uso terapéutico , Glucemia/metabolismo , Corticosterona/metabolismo , Citocinas/metabolismo , Disbiosis/metabolismo , Disbiosis/microbiología , Disbiosis/psicología , Disbiosis/terapia , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/microbiología , Hiperglucemia/microbiología , Inflamación , Ratones , Estrés Psicológico/psicología , Estrés Psicológico/terapia
15.
Inflammation ; 44(6): 2448-2462, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34657991

RESUMEN

Accumulating evidence has shown that inflammation, the gut microbiota, and neurotransmitters are closely associated with the pathophysiology of depression. However, the links between the gut microbiota and neurotransmitter metabolism remain poorly understood. The present study aimed to investigate the neuroinflammatory reactions in chronic restraint stress (CRS)-induced depression and to delineate the potential links between the gut microbiota and neurotransmitter metabolism. C57BL/6 mice were subjected to chronic restraint stress for 5 weeks, followed by behavioural tests (the sucrose preference test, forced swim test, open field test, and elevated plus maze) and analysis. The results showed that CRS significantly increased interleukin-1 beta (IL-1ß), interleukin-2 (IL-2), interleukin-6 (IL-6), and tumour necrosis factor α (TNFα) levels and decreased brain-derived neurotrophic factor (BDNF) expression, accompanied by the activation of IkappaB-alpha-phosphorylation-nuclear factor kappa-B (IκBα-p-NF-κB) signalling in the mouse hippocampus. In addition, the neurotransmitter metabolomics results showed that CRS resulted in decreased levels of plasma 5-hydroxytryptamine (5-HT), dopamine (DA), and noradrenaline (NE) and their corresponding metabolites, and gut microbiota faecal metabolites with the 16S rRNA gene sequencing indicated that CRS caused marked microbiota dysbiosis in mice, with a significant increase in Helicobacter, Lactobacillus, and Oscillibacter and a decrease in Parabacteroides, Ruminococcus, and Prevotella. Notably, CRS-induced depressive behaviours and the disturbance of neurotransmitter metabolism and microbiota dysbiosis can be substantially restored by dexamethasone (DXMS) administration. Furthermore, a Pearson heatmap focusing on correlations between the microbiota, behaviours, and neurotransmitters showed that Helicobacter, Lactobacillus, and Oscillibacter were positively correlated with depressive behaviours but were negatively correlated with neurotransmitter metabolism, and Parabacteroides and Ruminococcus were negatively correlated with depressive behaviours but were positively correlated with neurotransmitter metabolism. Taken together, the results suggest that inflammation is involved in microbiota dysbiosis and the disturbance of neurotransmitter metabolism in CRS-induced depressive changes, and the delineation of the potential links between the microbiota and neurotransmitter metabolism will provide novel strategies for depression treatment.


Asunto(s)
Bacterias/metabolismo , Conducta Animal , Monoaminas Biogénicas/metabolismo , Eje Cerebro-Intestino , Encéfalo/metabolismo , Depresión/microbiología , Microbioma Gastrointestinal , Mediadores de Inflamación/metabolismo , Estrés Psicológico/microbiología , Animales , Bacterias/genética , Depresión/inmunología , Depresión/metabolismo , Depresión/psicología , Modelos Animales de Enfermedad , Disbiosis , Heces/microbiología , Preferencias Alimentarias , Locomoción , Masculino , Aprendizaje por Laberinto , Metabolómica , Ratones Endogámicos C57BL , Restricción Física , Ribotipificación , Estrés Psicológico/inmunología , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Natación
16.
Biomed Pharmacother ; 143: 112087, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34474339

RESUMEN

Stress during pregnancy is not only detrimental to a woman's own physical and mental health, but can also cause changes in the intrauterine environment and even have an impact on later growth and development, this study was designed to understand the changes of gut microbiota in the maternal and offspring caused by prenatal chronic stress, and to explore the regulatory effect of LBP on gut microbiota, and then to improve the emotional damage caused by prenatal chronic stress in the offspring. A rat model of prenatal chronic stress was made and used LBP to intervene by gavage. Fresh feces of offspring were collected, the concentration of microbial metabolites were tested by ELISA. Illumina MiSeqPE300 sequencing technology was used to determine the sequence of 16S rRNA V3-V4 of microorganisms. On the PND 42, the emotional function of offspring were tested by open-field test (OFT), sucrose preference test (SPT) and tail of suspend test (TST). Results indicated that stress factors increased the plasma corticosterone level of rats during pregnancy and they appeared depressive behaviors. The body weight of offspring during prenatal chronic stress was lower than the control group, and the plasma corticosterone level was increased. Prenatal chronic stress had a significant impact on emotional performance of the offspring on OFT, SPT and TST. Alpha diversity of gut microbiota and microbiota composition in offspring of prenatal chronic stress was attenuated and some relationships existed between these parameters. LBP treatment reduced offspring's plasma corticosterone level and improved their body weight, changed the emotional function, increased the diversity of gut microbiota. Collectively, these findings disclose that prenatal chronic stress not only causes emotional injury on the offspring, but also changes the gut microbiota of the mother and offspring; LBP may regulate the intestinal flora of the mother, then reducing the influence of stress factors on the emotional injury of offspring.


Asunto(s)
Bacterias/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Emociones/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Estrés Psicológico/tratamiento farmacológico , Síntomas Afectivos/etiología , Síntomas Afectivos/microbiología , Síntomas Afectivos/prevención & control , Síntomas Afectivos/psicología , Animales , Bacterias/crecimiento & desarrollo , Eje Cerebro-Intestino/efectos de los fármacos , Enfermedad Crónica , Modelos Animales de Enfermedad , Disbiosis , Femenino , Preferencias Alimentarias/efectos de los fármacos , Masculino , Prueba de Campo Abierto/efectos de los fármacos , Embarazo , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Estrés Psicológico/microbiología , Estrés Psicológico/psicología
17.
Sci Rep ; 11(1): 17119, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429482

RESUMEN

Feather pecking (FP) is a stress-induced neuropsychological disorder of birds. Intestinal dysbiosis and inflammation are common traits of these disorders. FP is, therefore, proposed to be a behavioral consequence of dysregulated communication between the gut and the brain. Probiotic bacteria are known to favorably modulate the gut microbiome and hence the neurochemical and immune components of the gut-brain axis. Consequently, probiotic supplementation represents a promising new therapeutic to mitigate widespread FP in domestic chickens. We monitored FP, gut microbiota composition, immune markers, and amino acids related to the production of neurochemicals in chickens supplemented with Lactobacillus rhamnosus or a placebo. Data demonstrate that, when stressed, the incidence of FP increased significantly; however, L. rhamnosus prevented this increase. L. rhamnosus supplementation showed a strong immunological effect by increasing the regulatory T cell population of the spleen and the cecal tonsils, in addition to limiting cecal microbiota dysbiosis. Despite minimal changes in aromatic amino acid levels, data suggest that catecholaminergic circuits may be an interesting target for further studies. Overall, our findings provide the first data supporting the use of a single-strain probiotic to reduce stress-induced FP in chickens and promise to improve domestic birds' welfare.


Asunto(s)
Pollos/microbiología , Lacticaseibacillus rhamnosus/patogenicidad , Enfermedades de las Aves de Corral/microbiología , Probióticos/uso terapéutico , Estrés Psicológico/microbiología , Animales , Catecolaminas/metabolismo , Pollos/fisiología , Microbioma Gastrointestinal , Tonsila Palatina/inmunología , Enfermedades de las Aves de Corral/terapia , Probióticos/administración & dosificación , Bazo/inmunología , Estrés Psicológico/terapia
18.
Life Sci ; 282: 119821, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34271059

RESUMEN

AIMS: C57BL/6J mice are well-known to exhibit resilience to chronic social defeat stress (CSDS) for induction of depressive-like behavior. Establishment of protocols for reproducible induction of depressive-like behavior in C57BL/6J mice would be useful to elucidate the underlying molecular mechanisms using target gene-knock-in and -out mice whose background is generally C57BL/6J. Here, we developed a modified CSDS protocol for reproducible induction of depressive-like behavior in C57BL/6J mice, and compared the profile of their gut microbiota with that with the standard CSDS protocol. MAIN METHODS: To prevent acclimation of defeated C57BL/6J mice to aggressive ICR mice, the sensory contact following a daily 10 min-defeat episode was performed by housing an individual defeated mouse in a cage set next to a cage for the aggressor one. KEY FINDINGS: The number of attacks by ICR mice on C57BL/6J ones was significantly increased with the modified CSDS protocol, and the susceptible mice exhibited greater hippocampal inflammation and an increased immobility time in the forced swim test, compared in the case of the standard CSDS protocol, and the reproducibility was confirmed in another set of experiments. Both the standard and modified CSDS protocols changed the diversity and relative composition of gut microbiota in the susceptible mice, but there was no apparent difference in them between the standard and modified CSDS-susceptible mice. SIGNIFICANCE: We established a CSDS protocol for reproducible induction of depressive-like behavior in C57BL/6J mice, and the features of the gut microbiota were similar in the susceptible mice with and without the depressive-like behavior.


Asunto(s)
Conducta Animal , Depresión/microbiología , Microbioma Gastrointestinal , Derrota Social , Estrés Psicológico/microbiología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos ICR
19.
Behav Brain Res ; 414: 113474, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34280457

RESUMEN

Research has established that stress "gets under the skin," impacting neuroendocrine and neuroimmune pathways to influence risk for physical and mental health outcomes. These effects can be particularly significant for early life stress (ELS), or adverse childhood experiences (ACEs). In this review, we explore whether stress gets "into the belly," that is, whether psychosocial stress affects the gut microbiome. We review animal and human research utilizing a variety of stress paradigms (acute laboratory stressors, chronic stress, stressful life events, perceived stress, ELS, in utero stress) and their impacts on the gut microbiota, with a particular focus on ELS. We also review data on dietary interventions to moderate impact of stress on the gut microbiome. Our review suggests strong evidence that acute laboratory stress, chronic stress, and ELS affect the gut microbiota in rodents, and growing evidence that perceived stress and ELS may impact the gut microbiota in humans. Emerging data also suggests, particularly in rodents, that dietary interventions such as omega-3 fatty acids and pre- and pro-biotics may buffer against the effects of stress on the gut microbiome, but more research is needed. In sum, growing evidence suggests that stress impacts not only the neuroendocrine and neuroimmune axes, but also the microbiota-gut-brain-axis, providing a pathway by which stress may get "into the belly" to influence health risk.


Asunto(s)
Experiencias Adversas de la Infancia , Disbiosis , Microbioma Gastrointestinal , Estrés Psicológico , Animales , Disbiosis/dietoterapia , Disbiosis/etiología , Disbiosis/microbiología , Humanos , Estrés Psicológico/complicaciones , Estrés Psicológico/microbiología
20.
Behav Brain Res ; 414: 113471, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34280459

RESUMEN

The aromatic amino acid tryptophan (Trp) is a precursor for multiple metabolites that can steer proper immune and neurodevelopment as well as social behavior in later life. Dysregulation in the Trp metabolic pathways and abundance of Trp or its derivatives, including indoles, kynurenine (Kyn), and particularly serotonin, has been associated with behavioral deficits and neuropsychiatric disorders including autism spectrum disorders (ASD) and schizophrenia. Previously, we have shown that prenatal stress (PNS) alters placental Trp and serotonin, and reduces Trp-metabolizing members of the maternal colonic microbiota. Given that PNS also results in alterations in offspring neurodevelopment, behavior and immune function, we hypothesized that PNS affects Trp metabolism and transport in both the maternal and fetal compartments, and that these alterations continue into adolescence. We surmised that this is due to reductions in Trp-metabolizing microbes that would otherwise reduce the Trp pool under normal metabolic conditions. To test this, pregnant mice were exposed to a restraint stressor and gene expression of enzymes involved in Trp and serotonin metabolism were measured. Specifically, tryptophan 2,3-dioxygenase, aryl hydrocarbon receptor, and solute carrier proteins, were altered due to PNS both prenatally and postnatally. Additionally, Parasutterella and Bifidobacterium, which metabolize Trp in the gut, were reduced in both the dam and the offspring. Together, the reductions of Trp-associated microbes and concomitant dysregulation in Trp metabolic machinery in dam and offspring suggest that PNS-induced Trp metabolic dysfunction may mediate aberrant fetal neurodevelopment.


Asunto(s)
Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/microbiología , Estrés Psicológico/metabolismo , Estrés Psicológico/microbiología , Triptófano/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA