Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 193
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1324794, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39015337

RESUMO

Background: Despite mounting evidence of gut-brain involvement in psychiatric conditions, functional data remain limited, and analyses of other microbial niches, such as the vaginal microbiota, are lacking in relation to mental health. This aim of this study was to investigate if the connections between the gut microbiome and mental health observed in populations with a clinical diagnosis of mental illness extend to healthy women experiencing stress and depressive symptoms. Additionally, this study examined the functional pathways of the gut microbiota according to the levels of psychological symptoms. Furthermore, the study aimed to explore potential correlations between the vaginal microbiome and mental health parameters in young women without psychiatric diagnoses. Methods: In this cross-sectional study, 160 healthy Danish women (aged 18-40 years) filled out questionnaires with validated scales measuring symptoms of stress and depression and frequency of dietary intake. Fecal and vaginal microbiota samples were collected at the beginning of the menstrual cycle and vaginal samples were also collected at cycle day 8-12 and 18-22. Shotgun metagenomic profiling of the gut and vaginal microbiome was performed. The Kyoto Encyclopedia of Genes and Genomes (KEGG) was used for functional profiling and 56 Gut Brain Modules were analyzed in the fecal samples. Results: The relative abundance in the gut of the genera Escherichia, Parabacteroides, and Shigella was higher in women with elevated depressive symptoms. Women with high perceived stress showed a tendency of increased abundance of Escherichia, Shigella, and Blautia. Amongst others, the potentially pathogenic genera, Escherichia and Shigella correlate with alterations in the neuroactive pathways such as the glutamatergic, GABAeric, dopaminergic, and Kynurenine pathways. Vaginosis symptoms were more prevalent in women reporting high levels of stress and depressive symptoms. Conclusions: The findings of this study support the concept of a microbiota-associated effect on the neuroactive pathways even in healthy young women. This suggest, that targeting the gut microbiome could be a promising approach for future psychiatric interventions.


Assuntos
Depressão , Fezes , Microbioma Gastrointestinal , Estresse Psicológico , Vagina , Humanos , Feminino , Adulto , Adulto Jovem , Estudos Transversais , Adolescente , Depressão/microbiologia , Vagina/microbiologia , Fezes/microbiologia , Estresse Psicológico/microbiologia , Microbiota , Dinamarca , Voluntários Saudáveis , Eixo Encéfalo-Intestino/fisiologia , Inquéritos e Questionários , Metagenômica/métodos , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação
2.
Sci Rep ; 14(1): 15841, 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38982178

RESUMO

Intense psychosocial stress during early life has a detrimental effect on health-disease balance in later life. Simultaneously, despite its sensitivity to stress, the developing microbiome contributes to long-term health. Following stress exposure, HPA-axis activation regulates the "fight or flight" response with the release of glucose and cortisol. Here, we investigated the interaction between the oral microbiome and the stress response. We used a cohort of 115 adults, mean age 24, who either experienced institutionalisation and adoption (n = 40) or were non-adopted controls (n = 75). Glucose and cortisol measurements were taken from participants following an extended socially evaluated cold pressor test (seCPT) at multiple time points. The cohort´s oral microbiome was profiled via 16S-V4 sequencing on microbial DNA from saliva and buccal samples. Using mixed-effect linear regressions, we identified 12 genera that exhibited an interaction with host's cortisol-glucose response to stress, strongly influencing intensity and clearance of cortisol and glucose following stress exposure. Particularly, the identified taxa influenced the glucose and cortisol release profiles and kinetics following seCPT exposure. In conclusion, our study provided evidence for the oral microbiome modifying the effect of stress on the HPA-axis and human metabolism, as shown in glucose-cortisol time series data.


Assuntos
Hidrocortisona , Sistema Hipotálamo-Hipofisário , Microbiota , Sistema Hipófise-Suprarrenal , Saliva , Estresse Psicológico , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Estresse Psicológico/microbiologia , Estresse Psicológico/metabolismo , Hidrocortisona/metabolismo , Hidrocortisona/análise , Masculino , Feminino , Adulto , Sistema Hipófise-Suprarrenal/metabolismo , Saliva/microbiologia , Saliva/metabolismo , Adulto Jovem , Boca/microbiologia , Glucose/metabolismo
3.
PLoS One ; 19(7): e0306638, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38959280

RESUMO

Evidence supports that people identifying as a sexual or gender minority (SGMs) experience minority-related stress resulting from discrimination or expectations of prejudice, and that this is associated with increased mental and physical health problems compared to cisgender heterosexuals. However, the biological mechanisms driving minority-related stress impacts remain unknown, including the role of the gut microbiome. Thus, the aim of this study was to determine the relationship between SGM status and gut microbiome health among young adults attending a 4-year university. To this end, a prospective pilot study was completed in the fall and spring semesters of 2021-22. Self-identified SGMs (N = 22) and cisgender-heterosexuals (CIS-HET, N = 43) completed in-person interviews to provide mental health data and demographic information. Nail and saliva samples were collected at the time of interview to quantify chronic and acute cortisol. Stool samples were collected within 48 hours of interview for microbiome analysis. Assessment of the gut microbiota identified a significant reduction in alpha diversity among the SGM group, even when adjusting for mental health outcome. SGM group showed trends for higher abundance of microbes in phylum Bacteroidetes and lower abundance of microbes in phyla Firmicutes, Actinobacteria, and Proteobacteria compared to the CIS-HET group. These findings support that the gut microbiome could be contributing to negative health effects among the SGM community.


Assuntos
Microbioma Gastrointestinal , Minorias Sexuais e de Gênero , Humanos , Masculino , Feminino , Projetos Piloto , Minorias Sexuais e de Gênero/psicologia , Adulto Jovem , Adulto , Estudos Prospectivos , Fezes/microbiologia , Hidrocortisona/análise , Hidrocortisona/metabolismo , Saliva/microbiologia , Adolescente , Estresse Psicológico/microbiologia
4.
PLoS One ; 19(6): e0304787, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38837966

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Leite Humano , Estresse Psicológico , Humanos , Leite Humano/microbiologia , Feminino , Gravidez , Estresse Psicológico/microbiologia , Lactente , Recém-Nascido , Aleitamento Materno , Mães/psicologia
5.
Nutrients ; 16(11)2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38892694

RESUMO

Certain workplaces, like deep-sea voyages, subject workers to chronic psychological stress and circadian rhythm disorders due to confined environments and frequent shifts. In this study, participants lived in a strictly controlled confined environment, and we analyzed the effects of a confined environment on gut microbiota and metabolites. The results showed that living in confined environments can significantly alter both the gut microbiota and the gut metabolome, particularly affecting lipid metabolism pathways like glycerophospholipid metabolism. There was a significant reduction in the abundance of Faecalibacterium and Bacteroides, while Blautia, Bifidobacterium, and Collinsella showed significant increases. An association analysis revealed a strong correlation between changes in the gut microbiota and the metabolome. Four upregulated lipid metabolites may serve as biomarkers for damage induced by confined environments, and certain gut microbiota alterations, such as those involving Faecalibacterium and Bacteroides, could be potential psychobiotics or therapeutic targets for enhancing mental health in a confined environment.


Assuntos
Microbioma Gastrointestinal , Metaboloma , Humanos , Microbioma Gastrointestinal/fisiologia , Masculino , Adulto , Metabolismo dos Lipídeos , Bacteroides/metabolismo , Feminino , Estresse Psicológico/microbiologia , Estresse Psicológico/metabolismo , Fezes/microbiologia , Bactérias/metabolismo , Bactérias/classificação
6.
Mech Ageing Dev ; 220: 111956, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38906383

RESUMO

Psychological stress is a major contributing factor to several health problems (e.g., depression, cardiovascular disease). Around 35 % of the world's population suffers from it, including younger generations. Physiologically, stress manifests through neuroendocrine pathways (Hypothalamic-Pituitary-Adrenal (HPA) axis and Sympathetic-Adrenal-Medullary (SAM) system) which culminate in the production of stress mediators like cortisol, epinephrine and norepinephrine. Stress and its mediators have been associated to body aging, through molecular mechanisms such as telomere attrition, mitochondrial dysfunction, cellular senescence, chronic inflammation, and dysbiosis, among others. Regarding its impact in the skin, stress impacts its structural integrity and physiological function. Despite this review focusing on several hallmarks of aging, emphasis was placed on skin microbiota dysbiosis. In this line, several studies, comprising different age groups, demographic contexts and body sites, have reported skin microbiota alterations associated with aging, and some effects of stress mediators on skin microbiota have also been reviewed in this paper. From a different perspective, since it is not a "traditional" stress mediator, oxytocin, a cortisol antagonist, has been related to glucorticoids inhibition and to display positive effects on cellular aging. This hormone dysregulation has been associated to psychological issues such as depression, whereas its upregulation has been linked to positive social interaction.


Assuntos
Envelhecimento , Microbiota , Pele , Estresse Psicológico , Humanos , Pele/microbiologia , Pele/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Envelhecimento/metabolismo , Envelhecimento/fisiologia , Microbiota/fisiologia , Ocitocina/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Senescência Celular/fisiologia , Disbiose/microbiologia , Disbiose/metabolismo , Animais , Sistema Hipófise-Suprarrenal/metabolismo
7.
Commun Biol ; 7(1): 670, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822061

RESUMO

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.


Assuntos
Fezes , Microbioma Gastrointestinal , Estresse Psicológico , Animais , Fezes/microbiologia , Camundongos , Estresse Psicológico/microbiologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Bactérias/genética , Bactérias/classificação
8.
Front Immunol ; 15: 1365871, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38756771

RESUMO

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.


Assuntos
Eixo Encéfalo-Intestino , Encéfalo , Disbiose , Microbioma Gastrointestinal , Transtornos Mentais , Saúde Mental , Estresse Psicológico , Humanos , Microbioma Gastrointestinal/imunologia , Eixo Encéfalo-Intestino/imunologia , Estresse Psicológico/imunologia , Estresse Psicológico/microbiologia , Disbiose/imunologia , Transtornos Mentais/imunologia , Transtornos Mentais/microbiologia , Encéfalo/imunologia , Animais , Neuroimunomodulação
9.
Pharmacol Res ; 204: 107214, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38763328

RESUMO

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.


Assuntos
Depressão , Disbiose , Estresse Psicológico , Animais , Disbiose/metabolismo , Depressão/metabolismo , Depressão/microbiologia , Depressão/psicologia , Depressão/etiologia , Masculino , Humanos , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Estresse Psicológico/psicologia , Feminino , Adulto , Camundongos , Restrição Física/psicologia , Camundongos Endogâmicos C57BL , Microbioma Gastrointestinal , Eixo Encéfalo-Intestino , Boca/microbiologia , Pessoa de Meia-Idade , Saliva/metabolismo , Saliva/microbiologia , Comportamento Animal , Barreira Hematoencefálica/metabolismo
10.
Brain Behav Immun ; 119: 665-680, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38579936

RESUMO

Depression is a prevalent psychological condition with limited treatment options. While its etiology is multifactorial, both chronic stress and changes in microbiome composition are associated with disease pathology. Stress is known to induce microbiome dysbiosis, defined here as a change in microbial composition associated with a pathological condition. This state of dysbiosis is known to feedback on depressive symptoms. While studies have demonstrated that targeted restoration of the microbiome can alleviate depressive-like symptoms in mice, translating these findings to human patients has proven challenging due to the complexity of the human microbiome. As such, there is an urgent need to identify factors upstream of microbial dysbiosis. Here we investigate the role of mucin 13 as an upstream mediator of microbiome composition changes in the context of stress. Using a model of chronic stress, we show that the glycocalyx protein, mucin 13, is selectively reduced after psychological stress exposure. We further demonstrate that the reduction of Muc13 is mediated by the Hnf4 transcription factor family. Finally, we determine that deleting Muc13 is sufficient to drive microbiome shifts and despair behaviors. These findings shed light on the mechanisms behind stress-induced microbial changes and reveal a novel regulator of mucin 13 expression.


Assuntos
Depressão , Disbiose , Microbioma Gastrointestinal , Estresse Psicológico , Animais , Masculino , Camundongos , Comportamento Animal/fisiologia , Depressão/metabolismo , Depressão/microbiologia , Disbiose/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Fator 4 Nuclear de Hepatócito/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia
11.
Clin Transl Oncol ; 26(6): 1407-1418, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38194019

RESUMO

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.


Assuntos
Aminoácidos , Neoplasias da Mama , Depressão , Progressão da Doença , Microbioma Gastrointestinal , Neurotransmissores , Animais , Microbioma Gastrointestinal/fisiologia , Feminino , Camundongos , Neurotransmissores/metabolismo , Aminoácidos/metabolismo , Depressão/metabolismo , Depressão/microbiologia , Neoplasias da Mama/patologia , Neoplasias da Mama/microbiologia , Neoplasias da Mama/metabolismo , Metabolômica , Modelos Animais de Doenças , Estresse Psicológico/microbiologia , Estresse Psicológico/metabolismo , Estresse Psicológico/complicações
12.
Cell Metab ; 36(3): 466-483.e7, 2024 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-38266651

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Humanos , Linhagem da Célula , Estresse Psicológico/microbiologia , Acetatos , Indóis/farmacologia
14.
Dev Psychobiol ; 65(7): e22424, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37860905

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Mães , Feminino , Criança , Gravidez , Animais , Humanos , Lactente , Estresse Psicológico/microbiologia , Ansiedade , Transtornos de Ansiedade
15.
Front Cell Infect Microbiol ; 13: 1121445, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36909723

RESUMO

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.


Assuntos
Depressão , Microbioma Gastrointestinal , Camundongos , Masculino , Animais , Depressão/metabolismo , Depressão/microbiologia , Triptofano , Derrota Social , Camundongos Endogâmicos C57BL , Encéfalo , Bactérias , Estresse Psicológico/microbiologia
16.
Food Res Int ; 157: 111289, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35761597

RESUMO

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.


Assuntos
Depressão , Triptofano , Animais , Ansiedade , Comportamento Animal , Eixo Encéfalo-Intestino , Depressão/metabolismo , Dieta , Camundongos , Serotonina , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia
17.
Psychoneuroendocrinology ; 136: 105620, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34896741

RESUMO

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.


Assuntos
Antibacterianos , Depressão , Animais , Antibacterianos/farmacologia , Ansiedade/tratamento farmacológico , Comportamento Animal , Depressão/tratamento farmacológico , Depressão/microbiologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley , Serotonina , Estresse Psicológico/complicações , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/microbiologia
18.
PLoS One ; 16(12): e0260813, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34890405

RESUMO

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.


Assuntos
Indígena Americano ou Nativo do Alasca/estatística & dados numéricos , Bactérias/classificação , RNA Ribossômico 16S/genética , Análise de Sequência de DNA/métodos , Estresse Psicológico/epidemiologia , Vaginose Bacteriana/epidemiologia , Adulto , Bactérias/genética , Bactérias/isolamento & purificação , DNA Bacteriano/genética , DNA Ribossômico/genética , Feminino , Humanos , Microbiota , Pessoa de Meia-Idade , Filogenia , Prevalência , Estresse Psicológico/microbiologia , Estados Unidos/etnologia , Vagina/microbiologia , Vaginose Bacteriana/microbiologia , Adulto Jovem
19.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-34884735

RESUMO

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.


Assuntos
Metaboloma , Microbiota , Estresse Psicológico/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/urina , Masculino , Ratos Wistar , Estresse Psicológico/microbiologia
20.
Gut Microbes ; 13(1): 2000275, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34856844

RESUMO

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.


Assuntos
Aglomeração/psicologia , Microbioma Gastrointestinal , Estresse Psicológico/metabolismo , Estresse Psicológico/microbiologia , Animais , Antibacterianos/uso terapêutico , Glicemia/metabolismo , Corticosterona/metabolismo , Citocinas/metabolismo , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/psicologia , Disbiose/terapia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Hiperglicemia/microbiologia , Inflamação , Camundongos , Estresse Psicológico/psicologia , Estresse Psicológico/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...