Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(30): e2201967119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858435

RESUMEN

Major depressive disorder (MDD) is a prevalent and devastating mental illness. To date, the diagnosis of MDD is largely dependent on clinical interviews and questionnaires and still lacks a reliable biomarker. DNA methylation has a stable and reversible nature and is likely associated with the course and therapeutic efficacy of complex diseases, which may play an important role in the etiology of a disease. Here, we identified and validated a DNA methylation biomarker for MDD from four independent cohorts of the Chinese Han population. First, we integrated the analysis of the DNA methylation microarray (n = 80) and RNA expression microarray data (n = 40) and identified BICD2 as the top-ranked gene. In the replication phase, we employed the Sequenom MassARRAY method to confirm the DNA hypermethylation change in a large sample size (n = 1,346) and used the methylation-sensitive restriction enzymes and a quantitative PCR approach (MSE-qPCR) and qPCR method to confirm the correlation between DNA hypermethylation and mRNA down-regulation of BICD2 (n = 60). The results were replicated in the peripheral blood of mice with depressive-like behaviors, while in the hippocampus of mice, Bicd2 showed DNA hypomethylation and mRNA/protein up-regulation. Hippocampal Bicd2 knockdown demonstrates antidepressant action in the chronic unpredictable mild stress (CUMS) mouse model of depression, which may be mediated by increased BDNF expression. Our study identified a potential DNA methylation biomarker and investigated its functional implications, which could be exploited to improve the diagnosis and treatment of MDD.


Asunto(s)
Metilación de ADN , Trastorno Depresivo Mayor , Hipocampo , Proteínas Asociadas a Microtúbulos , Animales , ADN/metabolismo , Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Marcadores Genéticos , Hipocampo/metabolismo , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , ARN Mensajero/metabolismo , Estrés Psicológico/genética
2.
Front Microbiol ; 14: 1296116, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38260877

RESUMEN

Introduction: The ketone body ß-hydroxybutyric acid (BHB) plays critical roles in cellular proliferation and metabolic fuel utilization; however, its effects on the rumen microbiota remain unknown. Methods: Here, three doses of BHB (low, medium, and high) were supplemented to early-weaned goat kids. Results: Compared with controls, the beneficial effects of BHB on growth and rumen development were observed in goats at 90 days of age (d). The low dose of dietary BHB increased the concentration of rumen acetate, propionate, and butyrate on d90. The sequencing results of the rumen microbiota revealed marked shifts in rumen microbial community structure after early-weaned goat kids consumed BHB for 2 months. The signature bacterial ASVs for each treatment were identified and were the main drivers contributing to microbial interactions in the rumen. The bacteria associated with rumen weight were also correlated with body weight. Some classified bacterial signatures, including Prevotella, Olsenella umbonate, and Roseburia faecis, were related to rumen volatile fatty acids and host development. Conclusion: Overall, dietary BHB altered rumen microbiota and environments in young goats, which contributed to rumen development and growth.

3.
Diabetes ; 70(8): 1780-1793, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33962999

RESUMEN

Moods and metabolism modulate each other. High comorbidity of depression and metabolic disorders, such as diabetes and obesity, poses a great challenge to treat such conditions. Here we report the therapeutic efficacy of brain-derived neurotrophic factor (BDNF) by gene transfer in the dorsal raphe nucleus (DRN) in a chronic unpredictable mild stress model (CUMS) of depression and models of diabetes and obesity. In CUMS, BDNF-expressing mice displayed antidepressant- and anxiolytic-like behaviors, which are associated with augmented serotonergic activity. Both in the diet-induced obesity model (DIO) and in db/db mice, BDNF ameliorated obesity and diabetes, which may be mediated by enhanced sympathetic activity not involving DRN serotonin. Chronic activation of DRN neurons via chemogenetic tools produced similar effects as BDNF in DIO mice. These results established the DRN as a key nexus in regulating depression-like behaviors and metabolism, which can be exploited to combat comorbid depression and metabolic disorders via BDNF gene transfer.


Asunto(s)
Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Depresión/metabolismo , Núcleo Dorsal del Rafe/metabolismo , Metabolismo Energético/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Técnicas de Transferencia de Gen , Ratones , Neuronas/metabolismo , Obesidad/metabolismo
4.
J Affect Disord ; 273: 453-461, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32560940

RESUMEN

BACKGROUND: Telomeres are nucleoprotein complexes located at the end of chromosomes. Previous studies have confirmed that telomere length is reduced in the peripheral blood of depression patients. However, studies regarding whether telomere length is altered in brain regions associated with depression are limited. It remains unclear whether the peripheral blood telomere length indicates telomere variation in the brain. METHODS: Using quantitative PCR, we measured telomere length in five brain regions (prefrontal cortex, amygdala, nucleus accumbens, paraventricular nucleus, and hippocampus) from depressive-like mice and in peripheral blood from depressive-like mice and major depressive disorder (MDD) patients. We also examined the expression of telomerase- and alternative lengthening of telomere (ALT)-related genes in the prefrontal cortex and amygdala of depressive-like mice. RESULTS: Telomeres were shortened in the peripheral blood of depressive-like mice and MDD patients, but were elongated in the prefrontal cortex and amygdala compared with healthy controls. We also observed that the expression of ALT-related genes increased in the prefrontal cortex and amygdala. LIMITATIONS: The amount of human sample was limited. The mechanism of telomere lengthening in the brain of depressive-like mice was not well explained. Mice and humans have inherently different telomere and telomere maintenance systems. CONCLUSION: These findings illustrate that the telomere length in the peripheral blood may not indicate the dynamics of telomere length in the brain. They offer a new perspective on variable telomere length in different brain regions affected in depression and provide a new basis for understanding the relationship between variable telomere length and MDD.


Asunto(s)
Trastorno Depresivo Mayor , Telomerasa , Animales , Encéfalo/metabolismo , Trastorno Depresivo Mayor/genética , Humanos , Ratones , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , Homeostasis del Telómero/genética
5.
Aging Dis ; 10(3): 611-625, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31165005

RESUMEN

Diabetes is a systemic disease that can cause brain damage such as synaptic impairments in the hippocampus, which is partly because of neuroinflammation induced by hyperglycemia. Brain-derived neurotrophic factor (BDNF) is essential in modulating neuroplasticity. Its role in anti-inflammation in diabetes is largely unknown. In the present study, we investigated the effects of BDNF overexpression on reducing neuroinflammation and the underlying mechanism in mice with type 1 diabetes induced by streptozotocin (STZ). Animals were stereotactically microinjected in the hippocampus with recombinant adeno-associated virus (AAV) expressing BDNF or EGFP. After virus infection, four groups of mice, the EGFP+STZ, BDNF+STZ, EGFP Control and BDNF Control groups, received STZ or vehicle treatment as indicated. Three weeks later brain tissues were collected. We found that BDNF overexpression in the hippocampus significantly rescued STZ-induced decreases in mRNA and protein expression of two synaptic plasticity markers, spinophilin and synaptophysin. More interestingly, BDNF inhibited hyperglycemia-induced microglial activation and reduced elevated levels of inflammatory factors (TNF-α, IL-6). BDNF blocked the increase in HMGB1 levels and specifically, in levels of one of the HMGB1 receptors, RAGE. Downstream of HMGB1/RAGE, the increase in the protein level of phosphorylated NF-κB was also reversed by BDNF in STZ-treated mice. These results show that BDNF overexpression reduces neuroinflammation in the hippocampus of type 1 diabetic mice and suggest that the HMGB1/RAGE/NF-κB signaling pathway may contribute to alleviation of neuroinflammation by BDNF in diabetic mice.

6.
Neuron ; 100(3): 551-563.e7, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30220511

RESUMEN

Astrocyte dysfunction and inflammation are associated with the pathogenesis of major depressive disorder (MDD). However, the mechanisms underlying these effects remain largely unknown. Here, we found that multiple endocrine neoplasia type 1 (Men1; protein: menin) expression is attenuated in the brain of mice exposed to CUMS (chronic unpredictable mild stress) or lipopolysaccharide. Astrocyte-specific reduction of Men1 (GcKO) led to depressive-like behaviors in mice. We observed enhanced NF-κB activation and IL-1ß production with menin deficiency in astrocytes, where depressive-like behaviors in GcKO mice were restored by NF-κB inhibitor or IL-1ß receptor antagonist. Importantly, we identified a SNP, rs375804228, in human MEN1, where G503D substitution is associated with a higher risk of MDD onset. G503D substitution abolished menin-p65 interactions, thereby enhancing NF-κB activation and IL-1ß production. Our results reveal a distinct astroglial role for menin in regulating neuroinflammation in depression, indicating that menin may be an attractive therapeutic target in MDD.


Asunto(s)
Astrocitos/metabolismo , Trastorno Depresivo Mayor/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Estrés Psicológico/metabolismo , Adulto , Animales , Astrocitos/patología , Células Cultivadas , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/psicología , Femenino , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/genética , Estrés Psicológico/genética , Estrés Psicológico/psicología
7.
Gene ; 601: 21-26, 2017 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-27940106

RESUMEN

Genome-wide association (GWA) studies have identified numerous genetic variants for major depressive disorder (MDD) although most of the genetic variants are intergenic. It has been found that approximately 54% of long non-coding RNAs (lncRNAs) are located in the intergenic regions. We hypothesized that intergenic variants might be involved in the pathogenesis of MDD through regulating the expression of lncRNAs where these variants are located. In this study, several MDD-associated SNPs in three known intergenic lncRNAs were initially genotyped among 978 patients with MDD and 1176 controls, and the real-time PCR assay was performed to quantify the expression of LINC01108 and LINC00578 in peripheral blood cells from 20 MDD patients and 20 controls. The results showed that rs12526133 present in LINC01108 was strongly associated with MDD (χ2=11.68, P=6.3E-04), and LINC01108 expression was significantly higher in the patient group than in the control group (FC=1.90, P<0.001). The expression of LINC00998 was significantly lower in MDD patients than controls based on microarray analysis (FC=0.11, P<0.001), so that its tag SNPs were genotyped and rs2272260 in LINC00998 was found to be associated with MDD (χ2=26.39, P=2.8E-07). This work suggests that non-coding variants may play an important role in conferring risk of MDD.


Asunto(s)
ADN Intergénico/genética , Trastorno Depresivo Mayor/genética , ARN Largo no Codificante/genética , Adulto , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Adulto Joven
8.
Sci Rep ; 6: 37530, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27874048

RESUMEN

Long interspersed nuclear element-1 (LINE-1 or L1) is a type of retrotransposons comprising 17% of the human and mouse genome, and has been found to be associated with several types of neurological disorders. Previous post-mortem brain studies reveal increased L1 copy number in the prefrontal cortex from schizophrenia patients. However, whether L1 retrotransposition occurs similarly in major depressive disorder (MDD) is unknown. Here, L1 copy number was measured by quantitative PCR analysis in peripheral blood of MDD patients (n = 105) and healthy controls (n = 105). The results showed that L1 copy number was increased in MDD patients possibly due to its hypomethylation. Furthermore, L1 copy number in peripheral blood and five brain regions (prefrontal cortex, hippocampus, amygdala, nucleus accumbens and paraventricular hypothalamic nucleus) was measured in the chronic unpredictable mild stress (CUMS) model of depression in mice. Intriguingly, increased L1 copy number in blood and the decreased L1 copy number in the prefrontal cortex were observed in stressed mice, while no change was found in other brain regions. Our results suggest that the changes of L1 may be associated with the pathophysiology of MDD, but the biological mechanism behind dysfunction of L1 retrotransposition in MDD remains to be further investigated.


Asunto(s)
Encéfalo/metabolismo , Variaciones en el Número de Copia de ADN/genética , Trastorno Depresivo Mayor/genética , Elementos de Nucleótido Esparcido Largo/genética , Adulto , Encéfalo/patología , Metilación de ADN/genética , Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/fisiopatología , Femenino , Hipocampo , Humanos , Masculino , Persona de Mediana Edad , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Retroelementos/genética
9.
J Affect Disord ; 183: 279-86, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26047305

RESUMEN

BACKGROUND: Major depressive disorder (MDD) is a common, chronic and recurrent mental disease but the precise mechanism behind this disorder remains unknown. FTO is one of the N6-methyladenosine (m6A) modification genes and has recently been found to be associated with depression. N6-methyladenosine (m6A) is the most abundant internal modification on RNA, which is highly enriched within the brain. There are five genes involved in m6A modification including FTO, but whether these m6A modification genes could confer a risk of MDD is still unclear. This study aimed to investigate the genetic influence of the m6A modification genes on risk of MDD. METHODS: We genotyped 23 SNPs in 5 modification genes among 738 patients with MDD and 1098 controls. The UNPHASED program was applied to analyze the genotyping data for allelic and genotypic association with MDD. RESULTS: Of the 23 SNPs selected, rs12936694 from the m6A demethylase gene ALKBH5 showed allelic association (χ(2)=11.19, p=0.0008, OR=1.491, 95%CI 1.179-1.887) and genotypic association (χ(2)=12.26, df=2, p=0.0022) with MDD. LIMITATIONS: Replication and functional study are required to draw a firm conclusion. CONCLUSIONS: The ALKBH5 gene may play a role in conferring risk of MDD in the Chinese population.


Asunto(s)
Adenosina/análogos & derivados , Pueblo Asiatico/genética , Trastorno Depresivo Mayor/genética , Dioxigenasas/genética , Proteínas de la Membrana/genética , Adenosina/genética , Adulto , Desmetilasa de ARN, Homólogo 5 de AlkB , Alelos , Estudios de Casos y Controles , China/epidemiología , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA