Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Psychiatry ; 27(10): 4123-4135, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35444255

RESUMEN

The intricate processes of microbiota-gut-brain communication in modulating human cognition and emotion, especially in the context of mood disorders, have remained elusive. Here we performed faecal metagenomic, serum metabolomics and neuroimaging studies on a cohort of 109 unmedicated patients with depressed bipolar disorder (BD) patients and 40 healthy controls (HCs) to characterise the microbial-gut-brain axis in BD. Across over 12,000 measured metabolic features, we observed a large discrepancy (73.54%) in the serum metabolome between BD patients and HCs, spotting differentially abundant microbial-derived neuroactive metabolites including multiple B-vitamins, kynurenic acid, gamma-aminobutyric acid and short-chain fatty acids. These metabolites could be linked to the abundance of gut microbiota presented with corresponding biosynthetic potentials, including Akkermansia muciniphila, Citrobacter spp. (Citrobacter freundii and Citrobacter werkmanii), Phascolarctobacterium spp., Yersinia spp. (Yersinia frederiksenii and Yersinia aleksiciae), Enterobacter spp. (Enterobacter cloacae and Enterobacter kobei) and Flavobacterium spp. Based on functional neuroimaging, BD-related neuroactive microbes and metabolites were discovered as potential markers associated with BD-typical features of functional connectivity of brain networks, hinting at aberrant cognitive function, emotion regulation, and interoception. Our study combines gut microbiota and neuroactive metabolites with brain functional connectivity, thereby revealing potential signalling pathways from the microbiota to the gut and the brain, which may have a role in the pathophysiology of BD.


Asunto(s)
Trastorno Bipolar , Microbioma Gastrointestinal , Microbiota , Humanos , Microbioma Gastrointestinal/genética , Trastorno Bipolar/metabolismo , Eje Cerebro-Intestino , Metaboloma , Encéfalo/metabolismo
2.
J Cell Mol Med ; 25(14): 6463-6469, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34014031

RESUMEN

Bipolar disorder (BD) is a common psychiatric illness with high prevalence and disease burden. Accumulating susceptibility genes for BD have been identified in recent years. However, the exact functions of these genes remain largely unknown. Despite its high heritability, gene and environment interaction is commonly accepted as the major contributing factor to BD pathogenesis. Intestine microbiota is increasingly recognized as a critical environmental factor for human health and diseases via the microbiota-gut-brain axis. BD individuals showed altered diversity and compositions in the commensal microbiota. In addition to pro-inflammatory factors, such as interleukin-6 and tumour necrosis factor-α, type 1 interferon signalling pathway is also modulated by specific intestinal bacterial strains. Disruption of the microbiota-gut-brain axis contributes to peripheral and central nervous system inflammation, which accounts for the BD aetiology. Administration of type 1 interferon can induce the expression of TRANK1, which is associated with elevated circulating biomarkers of the impaired blood-brain barrier in BD patients. In this review, we focus on the influence of intestine microbiota on the expression of bipolar gene TRANK1 and propose that intestine microbiota-dependent type 1 interferon signalling is sufficient to induce the over-expression of TRANK1, consequently causing the compromise of BBB integrity and facilitating the entrance of inflammatory mediators into the brain. Activated neuroinflammation eventually contributes to the occurrence and development of BD. This review provides a new perspective on how gut microbiota participate in the pathogenesis of BD. Future studies are needed to validate these assumptions and develop new treatment targets for BD.


Asunto(s)
Trastorno Bipolar/genética , Eje Cerebro-Intestino/genética , Citocinas/genética , Microbioma Gastrointestinal/genética , Trastorno Bipolar/metabolismo , Trastorno Bipolar/microbiología , Trastorno Bipolar/patología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Susceptibilidad a Enfermedades , Humanos
3.
CNS Neurosci Ther ; 30(2): e14361, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37491837

RESUMEN

AIMS: We aimed to investigate whether peripheral T-cell subsets could be a biomarker to distinguish major depressive disorder (MDD) and bipolar disorder (BD). METHODS: Medical records of hospitalized patients in the Department of Psychiatry, the First Affiliated Hospital, Zhejiang University School of Medicine, from January 2015 to September 2020 with a discharge diagnosis of MDD or BD were reviewed. Patients who underwent peripheral blood examination of T-cell subtype proportions, including CD3+, CD4+, CD8+ T-cell, and natural killer (NK) cells, were enrolled. The Chi-square test, t-test, or one-way analysis of variance were used to analyze group differences. Demographic profiles and T-cell data were used to construct a random forest classifier-based diagnostic model. RESULTS: Totally, 98 cases of BD mania, 459 cases of BD depression (BD-D), and 458 cases of MDD were included. There were significant differences in the proportions of CD3+, CD4+, CD8+ T-cell, and NK cells among the three groups. Compared with MDD, the BD-D group showed higher CD8+ but lower CD4+ T-cell and a significantly lower ratio of CD4+ and CD8+ proportions. The random forest model achieved an area under the curve of 0.77 (95% confidence interval: 0.71-0.83) to distinguish BD-D from MDD patients. CONCLUSION: These findings imply that BD and MDD patients may harbor different T-cell inflammatory patterns, which could be a potential diagnostic biomarker for mood disorders.


Asunto(s)
Trastorno Bipolar , Trastorno Depresivo Mayor , Humanos , Trastorno Bipolar/diagnóstico , Trastorno Depresivo Mayor/diagnóstico , Estudios Retrospectivos , Subgrupos de Linfocitos T , Biomarcadores
4.
CNS Neurosci Ther ; 29 Suppl 1: 115-128, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36650644

RESUMEN

BACKGROUND: Most of the previous studies have demonstrated the potential antidepressive and anxiolytic role of prebiotic supplement in male subjects, yet few have females enrolled. Herein, we explored whether prebiotics administration during chronic stress prevented depression-like and anxiety-like behavior in a sex-specific manner and the mechanism of behavioral differences caused by sex. METHODS: Female and male C57 BL/J mice on normal diet were supplemented with or without a combination of fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS) during 3- and 4-week chronic restraint stress (CRS) treatment, respectively. C57 BL/J mice on normal diet without CRS were used as controls. Behavior consequences, gut microbiota, dysfunction of gut and brain-blood barriers, and inflammatory profiles were measured. RESULTS: In the 3rd week, FOS + GOS administration attenuated stress-induced anxiety-like behavior in female, but not in male mice, and the anxiolytic effects in males were observed until the 4th week. However, protective effects of prebiotics on CRS-induced depression were not observed. Changes in the gene expression of tight junction proteins in the distal colon and hippocampus, and decreased number of colon goblet cells following CRS were restored by prebiotics only in females. In both female and male mice, prebiotics alleviated stress-induced BBB dysfunction and elevation in pro-inflammatory cytokines levels, and modulated gut microbiota caused by stress. Furthermore, correlation analysis revealed that anxiety-like behaviors were significantly correlated with levels of pro-inflammatory cytokines and gene expression of tight junction proteins in the hippocampus of female mice, and the abundance of specific gut microbes was also correlated with anxiety-like behaviors, pro-inflammatory cytokines, and gene expression of tight junction proteins in the hippocampus of female mice. CONCLUSION: Female mice were more vulnerable to stress and prebiotics than males. The gut microbiota, gut and blood-brain barrier, and inflammatory response may mediate the protective effects of prebiotics on anxiety-like behaviors in female mice.


Asunto(s)
Ansiolíticos , Prebióticos , Femenino , Masculino , Ratones , Animales , Barrera Hematoencefálica/metabolismo , Depresión/etiología , Depresión/metabolismo , Caracteres Sexuales , Ansiedad/etiología , Citocinas/metabolismo , Oligosacáridos/farmacología
5.
J Affect Disord ; 324: 199-205, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36586603

RESUMEN

BACKGROUND: It is found that there are great differences in the efficacy of quetiapine at the same dose in many patients with bipolar disorders. Therefore, therapeutic drug monitoring (TDM) is a valuable tool for guiding treatment with quetiapine. The aims of this study were to assess the relationship between serum concentration and clinical response of quetiapine in adolescents and adults with bipolar disorders in acute stage. METHODS: The study design was prospective and observational. Within the naturalistic setting of a routine TDM service at the First Affiliated Hospital, Zhejiang University School of Medicine. Psychiatric symptoms were assessed using the HAMD (Hamilton Depression Scale), YRMS (Young manic rating scale) and CUDOS-M (Clinically Useful Depression Outcome Scale-Mixed Subscale). The decline of HAMD and YMRS scores was were used to assess clinical outcome of bipolar disorders respectively. RESULTS: 169 inpatients (23.7 % male, 76.3 % female) were enrolled in the study. We found that there was a strong correlation between quetiapine serum concentrations and clinical outcomes (rs = 0.702, p < 0.001). While, quetiapine daily dose was not correlated with clinical outcome. We found that when the quetiapine serum level is >146.85 ng/ml in depression episodes patients could obtain a satisfactory treatment effect after 2 weeks of hospitalization. CONCLUSIONS: We found a significant positive relationship between serum concentration and clinical outcome, and also determined the serum concentration of quetiapine for the treatment of bipolar depression.


Asunto(s)
Antipsicóticos , Trastorno Bipolar , Humanos , Masculino , Adulto , Femenino , Adolescente , Fumarato de Quetiapina/uso terapéutico , Trastorno Bipolar/psicología , Antipsicóticos/uso terapéutico , Estudios Prospectivos , Dibenzotiazepinas/uso terapéutico , Resultado del Tratamiento , Método Doble Ciego , Escalas de Valoración Psiquiátrica
6.
Front Psychiatry ; 12: 677039, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34381386

RESUMEN

Bipolar disorder (BD) is a severe affective disorder, mainly characterized by alternative depressive and manic or hypomanic episodes, yet the pathogenesis of BD has not been fully elucidated. Recent researches have implicated the altered kynurenine (KYN) metabolism involved in the neurobiology of BD. Excessive activation of the immune system also occurs in patients with BD, which further accelerates the KYN pathway for tryptophan metabolism. Changes of the KYN metabolites have effects on neuronal receptors and are involved in neuroendocrine transmissions. Interactions between KYN metabolism and the immune system may contribute to the neuropathogenesis of BD. Various studies have shown that alterations of the KYN metabolites were associated with mood, psychotic symptoms, and cognitive functions in patients with BD. In this review, we briefly introduce the KYN pathway and describe the immune dysregulation in BD as well as their interactions. We then focus on the research advances on the KYN metabolism in BD, which hold promise for identifying novel treatment targets in patients stricken with this disorder.

7.
Biosci Trends ; 15(5): 321-329, 2021 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-34588398

RESUMEN

Antipsychotic-induced metabolic dysfunction (AIMD) is an intractable clinical challenge worldwide. The situation is becoming more critical as second-generation antipsychotics (SGAs), to a great extent, have replaced the role of first-generation antipsychotics in managing major psychiatric disorders. Although the exact mechanisms for developing AIMD is intricate, emerging evidence has indicated the involvement of the microbiota-gut-brain axis in AIMD. SGAs treatment may change the diversity and compositions of intestinal flora (e.g., decreased abundance of Bacteroidetes and Akkermansia muciniphila, and increased Firmicutes). Short-chain fatty acids and other metabolites derived from gut microbiota, on the one hand, can regulate the activity of intestinal endocrine cells and their secretion of satiety hormones (e.g., glucagon-like peptide 1, peptide YY, cholecystokinin and ghrelin); on the other hand, can activate the vagus nerve or transport into the brain to exert a central modulation of foraging behaviors via binding to neuropeptide receptors. Interestingly, metformin, a classical antidiabetic agent, is capable of alleviating AIMD possibly by regulating the microbiota-gut-brain axis. That is, metformin can not only partially reverse the alterations of gut microbial communities due to SGAs treatment, but also play a positive role in rectifying the disturbances of peripheral and central satiety-related neuropeptides. Current evidence has indicated a promising role for metformin on ameliorating AMID, but further verifications in well-designed clinical trials are still warranted.


Asunto(s)
Antipsicóticos , Microbioma Gastrointestinal , Metformina , Antipsicóticos/efectos adversos , Encéfalo , Eje Cerebro-Intestino , Humanos , Metformina/uso terapéutico
8.
Front Immunol ; 12: 789647, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34992606

RESUMEN

Tetratricopeptide repeat and ankyrin repeat containing 1 (TRANK1) is a robust risk gene of bipolar disorder (BD). However, little is known on the role of TRANK1 in the pathogenesis of BD and whether the gut microbiota is capable of regulating TRANK1 expression. In this study, we first investigated the serum mRNA level of TRANK1 in medication-free patients with a depressive episode of BD, then a mice model was constructed by fecal microbiota transplantation (FMT) to explore the effects of gut microbiota on brain TRANK1 expression and neuroinflammation, which was further verified by in vitro Lipopolysaccharide (LPS) treatment in BV-2 microglial cells and neurons. 22 patients with a depressive episode and 28 healthy individuals were recruited. Serum level of TRANK1 mRNA was higher in depressed patients than that of healthy controls. Mice harboring 'BD microbiota' following FMT presented depression-like phenotype. mRNA levels of inflammatory cytokines and TRANK1 were elevated in mice hippocampus and prefrontal cortex. In vitro, LPS treatment activated the secretion of pro-inflammatory factors in BV-2 cells, which was capable of upregulating the neuronal expression of TRANK1 mRNA. Moreover, primary cortical neurons transfected with plasmid Cytomegalovirus DNA (pcDNA3.1(+)) vector encoding human TRANK1 showed decreased dendritic spine density. Together, these findings add new evidence to the microbiota-gut-brain regulation in BD, indicating that microbiota is possibly involved in the neuropathogenesis of BD by modulating the expression of TRANK1.


Asunto(s)
Trastorno Bipolar/inmunología , Eje Cerebro-Intestino/inmunología , Citocinas/metabolismo , Depresión/inmunología , Microbioma Gastrointestinal/inmunología , Adolescente , Adulto , Animales , Trastorno Bipolar/sangre , Trastorno Bipolar/microbiología , Trastorno Bipolar/patología , Estudios de Casos y Controles , Línea Celular , Citocinas/análisis , Depresión/sangre , Depresión/microbiología , Depresión/patología , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal , Femenino , Voluntarios Sanos , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Microglía/inmunología , Microglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Corteza Prefrontal/inmunología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Cultivo Primario de Células , Adulto Joven
9.
Clin Transl Med ; 10(4): e146, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32898322

RESUMEN

Trillions of microorganisms inhabiting in the human gut play an essential role in maintaining physical and mental health. The connections between gut microbiome and neuropsychiatric diseases have been recently identified. The pathogenesis of bipolar disorder, a spectrum of diseases manifesting with mood and energy fluctuations, also seems to be involved in the bidirectional modulation of the microbiome-gut-brain (MGB) axis. In this review, we briefly introduce the concept of MGB axis, and then focus on the previous findings in human studies associated with bipolar disorder. These studies provided preliminary evidences on the gut microbial alterations in bipolar disorder. Limitations in these studies and future directions in this research field, such as fecal microbiome transplantation and microbiome-targeted therapy, were discussed. A research framework linking gut microbiome to determinants and health-related outcomes in BD was also proposed. Better characterizing and understanding of gut microbial biosignatures in bipolar patients contribute to clarify the etiology of this intractable disease and pave the new way for treatment innovation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA