Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Neuroimmune Pharmacol ; 19(1): 36, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39042202

RESUMEN

Newly conducted research suggests that metabolic disorders, like diabetes and obesity, play a significant role as risk factors for psychiatric disorders. This connection presents a potential avenue for creating novel antidepressant medications by repurposing drugs originally developed to address antidiabetic conditions. Earlier investigations have shown that GLP-1 (Glucagon-like Peptide-1) analogs exhibit neuroprotective qualities in various models of neurological diseases, encompassing conditions such as Alzheimer's disease, Parkinson's disease, and stroke. Moreover, GLP-1 analogs have demonstrated the capability to enhance neurogenesis, a process recognized for its significance in memory formation and the cognitive and emotional aspects of information processing. Nonetheless, whether semaglutide holds efficacy as both an antidepressant and anxiolytic agent remains uncertain. To address this, our study focused on a mouse model of depression linked to type 2 diabetes induced by a High Fat Diet (HFD). In this model, we administered semaglutide (0.05 mg/Kg intraperitoneally) on a weekly basis to evaluate its potential as a therapeutic option for depression and anxiety. Diabetic mice had higher blood glucose, lipidic profile, and insulin resistance. Moreover, mice fed HFD showed higher serum interleukin (IL)-1ß and lipopolysaccharide (LPS) associated with impaired humor and cognition. The analysis of behavioral responses revealed that the administration of semaglutide effectively mitigated depressive- and anxiety-like behaviors, concurrently demonstrating an enhancement in cognitive function. Additionally, semaglutide treatment protected synaptic plasticity and reversed the hippocampal neuroinflammation induced by HFD fed, improving activation of the insulin pathway, demonstrating the protective effects of semaglutide. We also found that semaglutide treatment decreased astrogliosis and microgliosis in the dentate gyrus region of the hippocampus. In addition, semaglutide prevented the DM2-induced impairments of pro-opiomelanocortin (POMC), and G-protein-coupled receptor 43 (GPR43) and simultaneously increased the NeuN + and Glucagon-like Peptide-1 receptor (GLP-1R+) neurons in the hippocampus. Our data also showed that semaglutide increased the serotonin (5-HT) and serotonin transporter (5-HTT) and glutamatergic receptors in the hippocampus. At last, semaglutide changed the gut microbiota profile (increasing Bacterioidetes, Bacteroides acidifaciens, and Blautia coccoides) and decreased leaky gut, improving the gut-brain axis. Taken together, semaglutide has the potential to act as a therapeutic tool for depression and anxiety.


Asunto(s)
Ansiedad , Eje Cerebro-Intestino , Disfunción Cognitiva , Depresión , Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Péptidos Similares al Glucagón , Ratones Endogámicos C57BL , Animales , Péptidos Similares al Glucagón/farmacología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/psicología , Diabetes Mellitus Tipo 2/metabolismo , Ratones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/prevención & control , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Depresión/tratamiento farmacológico , Depresión/psicología , Depresión/metabolismo , Masculino , Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Ansiedad/etiología , Microbioma Gastrointestinal/efectos de los fármacos , Eje Cerebro-Intestino/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/psicología , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Antidepresivos/farmacología , Antidepresivos/uso terapéutico
2.
Sci Rep ; 14(1): 16215, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39003416

RESUMEN

The Apple polysaccharides (AP), extracted from the fruit of apple, has been used to treat multiple pathological diseases. In this study, we evaluated the effects of AP on cognitive impairment and intestinal aging in naturally aging mice. As a result, it was found that AP could improve spatial learning and memory impairment in aging mice through the Morris water maze experiment. Additionally, AP intervention can upregulate the expression of nerve growth factor (BDNF), postsynaptic marker (PSD95), and presynaptic marker (SYP) proteins. Moreover, AP can enhance total antioxidant capacity, reduce the level of pro-inflammatory cytokine, and inhibit the activation of the NF-κB signaling pathway, exerting anti-inflammatory and antioxidant functions. And the administration of AP restored intestinal mucosal barrier function, reduced the expression of aging and apoptosis related proteins. The administration of AP also altered the gut microbiota of mice. At the genus level, AP decreased the abundance of Helicobacter and Bilophila, while increased the abundance of Lactobacillus and Bacteroides. In summary, these data demonstrate that AP treatment can alleviate cognitive impairment, oxidative stress, and inflammatory reactions, repair the intestinal mucosal barrier, reduce intestinal aging, and alter specific microbial characteristics, ultimately improving the health of the elderly.


Asunto(s)
Envejecimiento , Eje Cerebro-Intestino , Disfunción Cognitiva , Microbioma Gastrointestinal , Malus , Polisacáridos , Animales , Polisacáridos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Malus/química , Ratones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Envejecimiento/efectos de los fármacos , Eje Cerebro-Intestino/efectos de los fármacos , Masculino , Estrés Oxidativo/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Intestinos/efectos de los fármacos , Intestinos/microbiología , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos
3.
J Neuroimmune Pharmacol ; 19(1): 37, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052165

RESUMEN

Recombinant interleukin-22 (rIL-22) has been reported as a protective agent in murine models of diseases driven by epithelial injury. Parasites have a circadian rhythm and their sensitivity to a certain drug may vary during the day. Therefore, this work aimed to investigate the effect of rIL-22 administration at different times of the day on the inflammation, oxidative status, and neurotransmitter release in the gut-brain axis of the Schistosoma mansoni-infected mice. Sixty male BALB/c mice aged six weeks weighing 25-30 g were divided into a control group (injected intraperitoneally with PBS), mice infected with 80 ± 10 cercariae of S. mansoni (infected group) then injected intraperitoneally with PBS, and rIL-22 treated groups. rIL-22 was administrated intraperitoneally (400 ng/kg) either at the onset or offset of the light phase for 14 days. IL-22 administration reduced the levels of IL-1ß, tumor necrosis factor-alpha (TNF-α), nuclear factor kappa beta (NF-κß), and enhanced the production of IL-22 and IL-17. The treatment with IL-22 increased glutathione (GSH) and reduced malondialdehyde (MDA) and nitric oxide (NO) levels both in the ileum and brain. The B-cell lymphoma 2 (BCL2) protein level in the ileum was diminished after IL-22 administration. Brain-derived neurotrophic factor (BDNF) and neurotransmitter release (serotonin, 5HT, norepinephrine, NE, dopamine, DA, Glutamate, Glu, and -amino butyric acid, GABA) were improved by rIL-22. In conclusion, rIL-22 showed promising immunotherapy for inflammation, oxidative damage, and neuropathological signs associated with schistosomiasis. The efficacy of IL-22 increased significantly upon its administration at the time of light offset.


Asunto(s)
Eje Cerebro-Intestino , Interleucina-22 , Interleucinas , Ratones Endogámicos BALB C , Neurotransmisores , Proteínas Recombinantes , Esquistosomiasis mansoni , Animales , Ratones , Masculino , Neurotransmisores/metabolismo , Neurotransmisores/farmacología , Interleucinas/metabolismo , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/administración & dosificación , Eje Cerebro-Intestino/efectos de los fármacos , Eje Cerebro-Intestino/fisiología , Inmunoterapia/métodos , Monoaminas Biogénicas/metabolismo , Inflamación/metabolismo , Inflamación/tratamiento farmacológico
4.
CNS Neurosci Ther ; 30(7): e14840, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38973202

RESUMEN

BACKGROUND: Heat stress (HS) commonly occurs as a severe pathological response when the body's sensible temperature exceeds its thermoregulatory capacity, leading to the development of chronic brain inflammation, known as neuroinflammation. Emerging evidence suggests that HS leads to the disruption of the gut microbiota, whereas abnormalities in the gut microbiota have been demonstrated to affect neuroinflammation. However, the mechanisms underlying the effects of HS on neuroinflammation are poorly studied. Meanwhile, effective interventions have been unclear. ß-Hydroxybutyric acid (BHBA) has been found to have neuroprotective and anti-inflammatory properties in previous studies. This study aims to explore the modulatory effects of BHBA on neuroinflammation induced by HS and elucidate the underlying molecular mechanisms. METHODS: An in vivo and in vitro model of HS was constructed under the precondition of BHBA pretreatment. The modulatory effects of BHBA on HS-induced neuroinflammation were explored and the underlying molecular mechanisms were elucidated by flow cytometry, WB, qPCR, immunofluorescence staining, DCFH-DA fluorescent probe assay, and 16S rRNA gene sequencing of colonic contents. RESULTS: Heat stress was found to cause gut microbiota disruption in HS mouse models, and TM7 and [Previotella] spp. may be the best potential biomarkers for assessing the occurrence of HS. Fecal microbiota transplantation associated with BHBA effectively reversed the disruption of gut microbiota in HS mice. Moreover, BHBA may inhibit microglia hyperactivation, suppress neuroinflammation (TNF-α, IL-1ß, and IL-6), and reduce the expression of cortical endoplasmic reticulum stress (ERS) markers (GRP78 and CHOP) mainly through its modulatory effects on the gut microbiota (TM7, Lactobacillus spp., Ruminalococcus spp., and Prevotella spp.). In vitro experiments revealed that BHBA (1 mM) raised the expression of the ERS marker GRP78, enhanced cellular activity, and increased the generation of reactive oxygen species (ROS) and anti-inflammatory cytokines (IL-10), while also inhibiting HS-induced apoptosis, ROS production, and excessive release of inflammatory cytokines (TNF-α and IL-1ß) in mouse BV2 cells. CONCLUSION: ß-Hydroxybutyric acid may be an effective agent for preventing neuroinflammation in HS mice, possibly due to its ability to inhibit ERS and subsequent microglia neuroinflammation via the gut-brain axis. These findings lay the groundwork for future research and development of BHBA as a preventive drug for HS and provide fresh insights into techniques for treating neurological illnesses by modifying the gut microbiota.


Asunto(s)
Ácido 3-Hidroxibutírico , Eje Cerebro-Intestino , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Enfermedades Neuroinflamatorias , Animales , Ratones , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/efectos de los fármacos , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Masculino , Ácido 3-Hidroxibutírico/farmacología , Trastornos de Estrés por Calor/metabolismo , Chaperón BiP del Retículo Endoplásmico , Fármacos Neuroprotectores/farmacología , Respuesta al Choque Térmico/fisiología , Respuesta al Choque Térmico/efectos de los fármacos
5.
Environ Toxicol Pharmacol ; 109: 104493, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908054

RESUMEN

The impacts of silicon dioxide nanoparticles (SiO2-NPs) on human health have attracted increasing interest due to their widespread utilization in medicine and food additives. However, the size-dependent effects of SiO2-NPs on brain health remain sparse. Herein we investigated alterations in behavioral patterns, the gut microbiota, inflammation and oxidative stress of mice after a 12-week exposure to SiO2-NPs with either small size (NP-S) or large size (NP-L). A more pronounced deleterious effect of NP-S was found on anxiety-like behavior in mice relative to NP-L. We also found that SiO2-NPs exposure induced inflammation and oxidative stress in the colon, hippocampus and cortex of mice in a size-specific manner. Correlation network analysis revealed potential links between anxiety-like behavior and SiO2-NPs-induced shifts in the gut microbiota including Parvibacter, Faecalibaculum, Gordonibacter and Ileibacterium. Furthermore, anxiety-like behavior caused by SiO2-NPs exposure exhibited correlations with decreased levels of hippocampal IL-10 and cortex Nqo1 as well as increased levels of intestinal Acox1 and hippocampal TNF-α. Therefore, our findings suggest that exposure to SiO2-NPs promoted anxiety-like behavior through the mediation of interplay between the gut and the brain, and SiO2-NPs of smaller size may generate a more adverse effect on brain health.


Asunto(s)
Ansiedad , Conducta Animal , Eje Cerebro-Intestino , Microbioma Gastrointestinal , Nanopartículas , Dióxido de Silicio , Animales , Dióxido de Silicio/toxicidad , Microbioma Gastrointestinal/efectos de los fármacos , Nanopartículas/toxicidad , Ansiedad/inducido químicamente , Masculino , Eje Cerebro-Intestino/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ratones Endogámicos C57BL
6.
J Affect Disord ; 360: 229-241, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38823591

RESUMEN

A high-fat diet can modify the composition of gut microbiota, resulting in dysbiosis. Changes in gut microbiota composition can lead to increased permeability of the gut barrier, allowing bacterial products like lipopolysaccharides (LPS) to enter circulation. This process can initiate systemic inflammation and contribute to neuroinflammation. Empagliflozin (EF), an SGLT2 inhibitor-type hypoglycemic drug, has been reported to treat neuroinflammation. However, there is a lack of evidence showing that EF regulates the gut microbiota axis to control neuroinflammation in HFD models. In this study, we explored whether EF could improve neuroinflammation caused by an HFD via regulation of the gut microbiota and the mechanism underlying this phenomenon. Our data revealed that EF alleviates pathological brain injury, reduces the reactive proliferation of astrocytes, and increases the expression of synaptophysin. In addition, the levels of inflammatory factors in hippocampal tissue were significantly decreased after EF intervention. Subsequently, the results of 16S rRNA gene sequencing showed that EF could change the microbial community structure of mice, indicating that the abundance of Lactococcus, Ligilactobacillus and other microbial populations decreased dramatically. Therefore, EF alleviates neuroinflammation by inhibiting gut microbiota-mediated astrocyte activation in the brains of high-fat diet-fed mice. Our study focused on the gut-brain axis, and broader research on neuroinflammation can provide a more holistic understanding of the mechanisms driving neurodegenerative diseases and inform the development of effective strategies to mitigate their impact on brain health. The results provide strong evidence supporting the larger clinical application of EF.


Asunto(s)
Astrocitos , Compuestos de Bencidrilo , Dieta Alta en Grasa , Microbioma Gastrointestinal , Glucósidos , Enfermedades Neuroinflamatorias , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Astrocitos/efectos de los fármacos , Glucósidos/farmacología , Ratones , Compuestos de Bencidrilo/farmacología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Masculino , Ratones Endogámicos C57BL , Encéfalo/efectos de los fármacos , Eje Cerebro-Intestino/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Disbiosis
7.
Benef Microbes ; 15(4): 373-385, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38897586

RESUMEN

Sleep quality and duration can be impacted by diet, and has been linked to gut microbiota composition and function as the result of communication via the microbiota-gut-brain axis. As one strategy to improve sleep quality could be through the modulation of the gut microbiome, we assessed the effects of a dairy-based product containing whey protein, galacto-oligosaccharides, tryptophan, vitamins and minerals after a 3 weeks intervention on gut microbiota composition and (gut-brain related) functions on basis of 67 healthy subjects with moderate sleep disturbances. Associations of the gut microbiota with sleep quality and with response/non-response to the treatment were revealed by shotgun metagenomics sequencing of faecal DNA samples, and subsequent analyses of microbiota taxonomy and generic functionality. A database of manually curated Gut-Brain Modules (GBMs) was applied to analyse specific microbial functions/pathways that have the potential to interact with the brain. A moderate discriminating effect of the DP treatment on gut microbiota composition was revealed which could be mainly attributed to a decrease in Pseudomonas resinovorans, Flintibacter sp. KGM00164, Intestinimonas butyriciproducens, and Flavonifractor plautii. As interindividual variance in microbiota composition could have given rise to a heterogenous responsiveness of the subjects in the intervention group, we zoomed in on the differences between responders and non-responders. A significant difference in baseline microbiota composition between responders and non-responders was apparent, showing lower Bifidobacterium longum and Bifidobacterium adolescentis, and higher Faecalibacterium prausnitzii relative abundances in responders. The findings provide leads with respect to the effectiveness and potential underlying mechanisms of mode of action in sleep improvement that could support future nutritional interventions to aid sleep improvement.


Asunto(s)
Productos Lácteos , Heces , Microbioma Gastrointestinal , Oligosacáridos , Calidad del Sueño , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Oligosacáridos/farmacología , Oligosacáridos/administración & dosificación , Adulto , Heces/microbiología , Femenino , Masculino , Productos Lácteos/microbiología , Persona de Mediana Edad , Bacterias/clasificación , Bacterias/genética , Bacterias/efectos de los fármacos , Bacterias/aislamiento & purificación , Metagenómica , Adulto Joven , Proteína de Suero de Leche/farmacología , Eje Cerebro-Intestino/efectos de los fármacos
8.
Sci Total Environ ; 945: 174026, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-38885706

RESUMEN

The rising global prevalence of microplastics (MPs) has highlighted their diverse toxicological effects. The oxytocin (OT) system in mammals, deeply intertwined with social behaviors, is recognized to be vulnerable to environmental stressors. We hypothesized that MP exposure might disrupt this system, a topic not extensively studied. We investigated the effects of MPs on behavioral neuroendocrinology via the gut-brain axis by exposing adolescent male C57BL/6 mice to varied sizes (5 µm and 50 µm) and concentrations (100 µg/L and 1000 µg/L) of polystyrene MPs over 10 weeks. The results demonstrated that exposure to 50 µm MPs significantly reduced colonic mucin production and induced substantial alterations in gut microbiota. Notably, the 50 µm-100 µg/L group showed a significant reduction in OT content within the medial prefrontal cortex and associated deficits in sociality, along with damage to the blood-brain barrier. Importantly, blocking the vagal pathway ameliorated these behavioral impairments, emphasizing the pivotal role of the gut-brain axis in mediating neurobehavioral outcomes. Our findings confirm the toxicity of MPs on sociality and the corresponding neuroendocrine systems, shedding light on the potential hazards and adverse effects of environmental MPs exposure on social behavior and neuroendocrine frameworks in social mammals, including humans.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Ratones Endogámicos C57BL , Microplásticos , Oxitocina , Poliestirenos , Conducta Social , Animales , Oxitocina/metabolismo , Ratones , Masculino , Poliestirenos/toxicidad , Microplásticos/toxicidad , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos
9.
Artículo en Inglés | MEDLINE | ID: mdl-38885875

RESUMEN

Mounting evidence points towards a crucial role of the kynurenine pathway (KP) in the altered gut-brain axis (GBA) balance in severe mental illness (SMI, namely depression, bipolar disorder, and schizophrenia) and cardiometabolic comorbidities. Preliminary evidence shows that serotonergic psychedelics and their analogues may hold therapeutic potential in addressing the altered KP in the dysregulated GBA in SMI and comorbidities. In fact, aside from their effects on mood, psychedelics elicit therapeutic improvement in preclinical models of obesity, metabolic syndrome, and vascular inflammation, which are highly comorbid with SMI. Here, we review the literature on the therapeutic modulation of the KP in the dysregulated GBA in SMI and comorbidities, and the potential application of psychedelics to address the altered KP in the brain and systemic dysfunction underlying SMI and comorbidities. Psychedelics might therapeutically modulate the KP in the altered GBA in SMI and comorbidities either directly, via altering the metabolic pathway by influencing the rate-limiting enzymes of the KP and affecting the levels of available tryptophan, or indirectly, by affecting the gut microbiome, gut metabolome, metabolism, and the immune system. Despite promising preliminary evidence, the mechanisms and outcomes of the KP modulation with psychedelics in SMI and systemic comorbidities remain largely unknown and require further investigation. Several concerns are discussed surrounding the potential side effects of this approach in specific cohorts of individuals with SMI and systemic comorbidities.


Asunto(s)
Alucinógenos , Quinurenina , Humanos , Quinurenina/metabolismo , Alucinógenos/uso terapéutico , Alucinógenos/farmacología , Animales , Eje Cerebro-Intestino/efectos de los fármacos , Eje Cerebro-Intestino/fisiología , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/metabolismo , Comorbilidad , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología
10.
Biochem Pharmacol ; 226: 116383, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908530

RESUMEN

The short-chain fatty acids (SCFAs) acetate, propionate and butyrate, the major products of intestinal microbial fermentation of dietary fibres, are involved in fine-tuning brain functions via the gut-brain axis. However, the effects of SCFAs in the hypothalamic neuronal network regulating several autonomic-brain functions are still unknown. Using NMR spectroscopy, we detected a reduction in brain acetate concentrations in the hypothalamus of obese leptin knockout ob/ob mice compared to lean wild-type littermates. Therefore, we investigated the effect of acetate on orexin/hypocretin neurons (hereafter referred as OX or OX-A neurons), a subset of hypothalamic neurons regulating energy homeostasis, which we have characterized in previous studies to be over-activated by the lack of leptin and enhancement of endocannabinoid tone in the hypothalamus of ob/ob mice. We found that acetate reduces food-intake in concomitance with a reduction of orexin neuronal activity in ob/ob mice. This was demonstrated by evaluating food-intake behaviour and orexin-A/c-FOS immunoreactivity coupled with patch-clamp recordings in Hcrt-eGFP neurons, quantification of prepro-orexin mRNA, and immunolabeling of GPR-43, the main acetate receptor. Our data provide new insights into the mechanisms of the effects of chronic dietary supplementation with acetate, or complex carbohydrates, on energy intake and body weight, which may be partly mediated by inhibition of orexinergic neuron activity.


Asunto(s)
Acetatos , Eje Cerebro-Intestino , Metabolismo Energético , Homeostasis , Neuronas , Orexinas , Animales , Orexinas/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Ratones , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/fisiología , Acetatos/farmacología , Acetatos/metabolismo , Eje Cerebro-Intestino/efectos de los fármacos , Eje Cerebro-Intestino/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/farmacología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Hipotálamo/metabolismo , Hipotálamo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología
11.
Artículo en Inglés | MEDLINE | ID: mdl-38914414

RESUMEN

One of the most challenging and controversial issues in microbiome research is related to gut microbial metabolism and neuropsychological disorders. Psychobiotics affect human behavior and central nervous system processes via the gut-brain axis, involving neuronal, immune, and metabolic pathways. They have therapeutic potential in the treatment of several neurodegenerative and neurodevelopmental disorders such as depression, anxiety, autism, attention deficit hyperactivity disorder, Alzheimer's disease, Parkinson's disease, schizophrenia, Huntington's disease, anorexia nervosa, and multiple sclerosis. However, the mechanisms underlying the interaction between psychobiotics and the abovementioned diseases need further exploration. This review focuses on the relationship between gut microbiota and its impact on neurological and neurodegenerative disorders, examining the potential of psychobiotics as a preventive and therapeutic approach, summarising recent research on the gut-brain axis and the potential beneficial effects of psychobiotics, highlighting the need for further research and investigation in this area.


Asunto(s)
Eje Cerebro-Intestino , Microbioma Gastrointestinal , Humanos , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Probióticos/uso terapéutico , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/terapia , Salud Mental , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico
12.
Toxicology ; 506: 153871, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38925359

RESUMEN

Fmr1 (fragile X messenger ribonucleoprotein 1)-knockout (KO) rats, modeling the human Fragile X Syndrome (FXS), are of particular interest for exploring the ASD-like phenotype in preclinical studies. Gestational exposure to chlorpyrifos (CPF) has been associated with ASD diagnosis in humans and ASD-like behaviors in rodents and linked to the microbiota-gut-brain axis. In this study, we have used both Fmr1-KO and wild-type male rats (F2 generation) at postnatal days (PND) 7 and 40 obtained after F1 pregnant females were randomly exposed to 1 mg/kg/mL/day of CPF or vehicle. A nuclear magnetic resonance (NMR) metabolomics approach together with gene expression profiles of these F2 generation rats were employed to analyze different brain regions (such as prefrontal cortex, hippocampus, and cerebellum), whole large intestine (at PND7) and gut content (PND40). The statistical comparison of each matrix spectral profile unveiled tissue-specific metabolic fingerprints. Significant variations in some biomarker levels were detected among brain tissues of different genotypes, including taurine, myo-inositol, and 3-hydroxybutyric acid, and exposure to CPF induced distinct metabolic alterations, particularly in serine and myo-inositol. Additionally, this study provides a set of metabolites associated with gastrointestinal dysfunction in ASD, encompassing several amino acids, choline-derived compounds, bile acids, and sterol molecules. In terms of gene expression, genotype and gestational exposure to CPF had only minimal effects on decarboxylase 2 (gad2) and cholinergic receptor muscarinic 2 (chrm2) genes.


Asunto(s)
Trastorno del Espectro Autista , Biomarcadores , Eje Cerebro-Intestino , Cloropirifos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Microbioma Gastrointestinal , Efectos Tardíos de la Exposición Prenatal , Animales , Cloropirifos/toxicidad , Embarazo , Femenino , Masculino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Microbioma Gastrointestinal/efectos de los fármacos , Biomarcadores/metabolismo , Trastorno del Espectro Autista/inducido químicamente , Trastorno del Espectro Autista/metabolismo , Eje Cerebro-Intestino/efectos de los fármacos , Ratas , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos
13.
Mar Drugs ; 22(6)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38921560

RESUMEN

Pinctada martensii hydrolysate (PMH) has been proved to have the effect of ameliorating disorders of glucose and lipid metabolism in db/db mice, but the mechanism of its hyperglycemia effect is still unclear. Bacterial communities in fecal samples from a normal control group, a diabetic control group, and a PMH-treated diabetes mellitus type 2 (T2DM) group were analyzed by 16S gene sequencing. Nano LC-MS/MS was used to analyze mice neuropeptides and proteomes. The 16S rDNA sequencing results showed that PMH modulated the structure and composition of the gut microbiota and improved the structure and composition of Firmicutes and Bacteroidetes at the phylum level and Desulfovibrionaceae and Erysipelatoclostridiaceae at the family level. Furthermore, the expressions of functional proteins of the central nervous system, immune response-related protein, and proteins related to fatty acid oxidation in the brain disrupted by an abnormal diet were recovered by PMH. PMH regulates the brain neuropeptidome and proteome and further regulates blood glucose in diabetic mice through the gut-brain axis. PMH may be used as a prebiotic agent to attenuate T2DM, and target-specific microbial species may have unique therapeutic promise for metabolic diseases.


Asunto(s)
Encéfalo , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Proteoma , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Masculino , Eje Cerebro-Intestino/efectos de los fármacos , Glucemia/efectos de los fármacos , Ratones Endogámicos C57BL , Prebióticos , Heces/microbiología , Heces/química
14.
Pharmacol Biochem Behav ; 241: 173805, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38848976

RESUMEN

Depression, a prevalent mental health condition, significantly impacts global mental impairment rates. While antidepressants are commonly used, treatment-resistant depression (TRD) poses a challenge. Emerging research highlights the role of the gut microbiota in depression through the gut-brain axis. This study identifies key genes associated with depression influenced by specific gut microbiota, Coprococcus and Subdoligranulum. Using bioinformatics tools, potential targets were elucidated, and molecular docking studies were performed. Furthermore, gene expression analysis identified hub-genes related to depression, intersecting with metabolite targets. Protein-protein interaction analysis revealed pivotal targets such as PTGS2 and MMP9. Molecular docking demonstrated 3-Indolepropionic acid's superior affinity over (R)-3-(4-Hydroxyphenyl)lactate. Physicochemical properties and toxicity profiles were compared, suggesting favorable attributes for 3-Indolepropionic acid. Molecular dynamics simulations confirmed stability and interactions of compounds with target proteins. This comprehensive approach sheds light on the complex interplay between gut microbiota, genes, and depression, emphasizing the potential for microbiota-targeted interventions in mental health management.


Asunto(s)
Microbioma Gastrointestinal , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Farmacología en Red , Indoles/farmacología , Depresión/tratamiento farmacológico , Depresión/microbiología , Depresión/metabolismo , Propionatos/farmacología , Propionatos/metabolismo , Eje Cerebro-Intestino/efectos de los fármacos , Antidepresivos/farmacología
15.
Mol Nutr Food Res ; 68(11): e2400090, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38757671

RESUMEN

SCOPE: Depression, a prevalent mental disorder, has significantly impacted the lives of 350 million people, yet it holds promise for amelioration through food-derived phenolics. Raspberries, renowned globally for their delectable flavor, harbor a phenolic compound known as raspberry ketone (RK). However, the impact of RK on depressive symptoms remains ambiguous. This study aims to investigate the impact of RK on lipopolysaccharide (LPS)-induced depressed mice and elucidates its potential mechanisms, focusing on the gut-brain axis. METHODS AND RESULTS: Through behavioral tests, RK exerts a notable preventive effect on LPS-induced depression-like behaviors in mice. RK proves capable of attenuating gut inflammation, repairing gut barrier impairment, modulating the composition of the gut microbiome (Muribaculaceae, Streptococcus, Lachnospiraceae, and Akkermansia), and promoting the production of short-chain fatty acids. Furthermore, RK alleviates neuroinflammation by suppressing the TLR-4/NF-κB pathway and bolsters synaptic function by elevating levels of neurotrophic factors and synapse-associated proteins. CONCLUSION: The current study provides compelling evidence that RK effectively inhibits the TLR-4/NF-κB pathway via the gut-brain axis, leading to the improvement of LPS-induced depression-like behaviors in mice. This study addresses the research gap in understanding the antidepressant effects of RK and illuminates the potential of utilizing RK as a functional food for preventing depression.


Asunto(s)
Eje Cerebro-Intestino , Depresión , Microbioma Gastrointestinal , Lipopolisacáridos , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Animales , Receptor Toll-Like 4/metabolismo , Lipopolisacáridos/toxicidad , FN-kappa B/metabolismo , Depresión/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Masculino , Ratones , Eje Cerebro-Intestino/efectos de los fármacos , Eje Cerebro-Intestino/fisiología , Butanonas/farmacología , Ratones Endogámicos C57BL , Conducta Animal/efectos de los fármacos , Antidepresivos/farmacología
16.
J Neuroinflammation ; 21(1): 124, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730498

RESUMEN

Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.


Asunto(s)
Antiinflamatorios , Lesiones Traumáticas del Encéfalo , Eje Cerebro-Intestino , Humanos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Eje Cerebro-Intestino/fisiología , Eje Cerebro-Intestino/efectos de los fármacos , Animales , Antiinflamatorios/uso terapéutico , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/etiología
17.
Nutrients ; 16(9)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38732599

RESUMEN

In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics-probiotics beneficial to mental health via the gut-brain axis-in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.


Asunto(s)
Trastornos Mentales , Probióticos , Ensayos Clínicos Controlados Aleatorios como Asunto , Humanos , Probióticos/uso terapéutico , Femenino , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/terapia , Trastornos del Conocimiento/tratamiento farmacológico , Masculino , Resultado del Tratamiento , Adulto , Eje Cerebro-Intestino/efectos de los fármacos , Persona de Mediana Edad , Microbioma Gastrointestinal/efectos de los fármacos , Lactobacillus , Anciano , Bifidobacterium
18.
Phytomedicine ; 129: 155510, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38696921

RESUMEN

BACKGROUND: Gut microbiota plays a critical role in the pathogenesis of depression and are a therapeutic target via maintaining the homeostasis of the host through the gut microbiota-brain axis (GMBA). A co-decoction of Lilii bulbus and Radix Rehmannia Recens (LBRD), in which verbascoside is the key active ingredient, improves brain and gastrointestinal function in patients with depression. However, in depression treatment using verbascoside or LBRD, mechanisms underlying the bidirectional communication between the intestine and brain via the GMBA are still unclear. PURPOSE: This study aimed to examine the role of verbascoside in alleviating depression via gut-brain bidirectional communication and to study the possible pathways involved in the GMBA. METHODS: Key molecules and compounds involved in antidepressant action were identified using HPLC and transcriptomic analyses. The antidepressant effects of LBRD and verbascoside were observed in chronic stress induced depression model by behavioural test, neuronal morphology, and synaptic dendrite ultrastructure, and their neuroprotective function was measured in corticosterone (CORT)-stimulated nerve cell injury model. The causal link between the gut microbiota and the LBRD and verbascoside antidepressant efficacy was evaluate via gut microbiota composition analysis and faecal microbiota transplantation (FMT). RESULTS: LBRD and Verbascoside administration ameliorated depression-like behaviours and synaptic damage by reversing gut microbiota disturbance and inhibiting inflammatory responses as the result of impaired intestinal permeability or blood-brain barrier leakiness. Furthermore, verbascoside exerted neuroprotective effects against CORT-induced cytotoxicity in an in vitro depression model. FMT therapy indicated that verbascoside treatment attenuated gut inflammation and central nervous system inflammatory responses, as well as eliminated neurotransmitter and brain-gut peptide deficiencies in the prefrontal cortex by modulating the composition of gut microbiota. Lactobacillus, Parabacteroides, Bifidobacterium, and Ruminococcus might play key roles in the antidepressant effects of LBRD via the GMBA. CONCLUSION: The current study elucidates the multi-component, multi-target, and multi-pathway therapeutic effects of LBRD on depression by remodeling GMBA homeostasis and further verifies the causality between gut microbiota and the antidepressant effects of verbascoside and LBRD.


Asunto(s)
Antidepresivos , Eje Cerebro-Intestino , Depresión , Microbioma Gastrointestinal , Glucósidos , Enfermedades Neuroinflamatorias , Fenoles , Rehmannia , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Rehmannia/química , Glucósidos/farmacología , Eje Cerebro-Intestino/efectos de los fármacos , Depresión/tratamiento farmacológico , Masculino , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Antidepresivos/farmacología , Fenoles/farmacología , Ratones , Estrés Psicológico/tratamiento farmacológico , Modelos Animales de Enfermedad , Permeabilidad , Ratas , Encéfalo/efectos de los fármacos , Ratones Endogámicos C57BL , Funcion de la Barrera Intestinal , Polifenoles
19.
J Hazard Mater ; 472: 134444, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38701724

RESUMEN

The effects of antipsychotic drugs on aquatic organisms have received widespread attention owing to their widespread use and continued release in aquatic environments. The toxicological effects of antipsychotics on aquatic organisms, particularly fish, are unexplored, and the underlying mechanisms remain unelucidated. This study aimed to use common carp to explore the effects of antipsychotics (olanzapine [OLA] and risperidone [RIS]) on behavior and the potential mechanisms driving these effects. The fish were exposed to OLA (0.1 and 10 µg/L) and RIS (0.03 and 3 µg/L) for 60 days. Behavioral tests and neurological indicators showed that exposure to antipsychotics could cause behavioral abnormalities and neurotoxicity in common carp. Further, 16 S rRNA sequencing revealed gut microbiota alteration and decreased relative abundance of some strains related to SCFA production after OLA and RIS exposure. Subsequently, a pseudo-sterile common carp model was successfully constructed, and transplantation of the gut microbiota from antipsychotic-exposed fish caused behavioral abnormalities and neurotoxicity in pseudo-sterile fish. Further, SCFA supplementation demonstrated that SCFAs ameliorated the behavioral abnormalities and neurological damage caused by antipsychotic exposure. To our knowledge, the present study is the first to investigate the effects of antipsychotics on various complex behaviors (swimming performance and social behavior) in common carp, highlighting the potential health risks associated with antipsychotic drug-induced neurotoxicity in fish. Although these results do not fully elucidate the mechanisms underlying the effects of antipsychotic drugs on fish behavior, they serve as a valuable initial investigation and form the basis for future research.


Asunto(s)
Antipsicóticos , Conducta Animal , Carpas , Microbioma Gastrointestinal , Risperidona , Contaminantes Químicos del Agua , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Antipsicóticos/toxicidad , Conducta Animal/efectos de los fármacos , Risperidona/toxicidad , Risperidona/farmacología , Contaminantes Químicos del Agua/toxicidad , Olanzapina/toxicidad , Eje Cerebro-Intestino/efectos de los fármacos , Natación , Conducta Social
20.
Phytomedicine ; 130: 155744, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38763011

RESUMEN

BACKGROUND: Aging is associated with learning and memory disorder, affecting multiple brain areas, especially the hippocampus. Previous studies have demonstrated trilobatin (TLB), as a natural food additive, can extend the life of Caenorhabditis elegans and exhibit neuroprotection in Alzheimer's disease mice. However, the possible significance of TLB in anti-aging remains elusive. PURPOSE: This study aimed to delve into the physiological mechanism by which TLB ameliorated aging-induced cognitive impairment in senescence-accelerated mouse prone 8 (SAMP8) mice. METHODS: 6-month-old SAMP8 mice were administrated with TLB (5, 10, 20 mg/kg/day, i.g.) for 3 months. The therapeutic effect of TLB on aging-induced cognitive impairment was assessed in mice using behavioral tests and aging score. The gut microbiota composition in fecal samples was analyzed by metagenomic analysis. The protective effects of TLB on blood-brain barrier (BBB) and intestinal barrier were detected by transmission electron microscope, H&E staining and western blot (WB) assay. The inhibitive effects of TLB on inflammation in brain and intestine were assessed using immunofluorescence, WB and ELISA assay. Molecular docking and surface plasma resonance (SPR) assay were utilized to investigate interaction between TLB and sirtuin 2 (SIRT2). RESULTS: Herein, the findings exhibited TLB mitigated aging-induced cognitive impairment, neuron injury and neuroinflammation in hippocampus of aged SAMP8 mice. Moreover, TLB treatment repaired imbalance of gut microbiota in aged SAMP8 mice. Furthermore, TLB alleviated the damage to BBB and intestinal barrier, concomitant with reducing the expression of SIRT2, phosphorylated levels of c-Jun NH2 terminal kinases (JNK) and c-Jun, and expression of MMP9 protein in aged SAMP8 mice. Molecular docking and SPR unveiled TLB combined with SIRT2 and down-regulated SIRT2 protein expression. Mechanistically, the potential mechanism of SIRT2 in TLB that exerted anti-aging effect was validated in vitro. As expected, SIRT2 deficiency attenuated phosphorylated level of JNK in HT22 cells treated with d-galactose. CONCLUSION: These findings reveal, for the first time, SIRT2-mediated brain-gut barriers contribute to aging and aging-related diseases, and TLB can rescue aging-induced cognitive impairment by targeting SIRT2 and restoring gut microbiota disturbance to mediate the brain-gut axis. Overall, this work extends the potential application of TLB as a natural food additive in aging-related diseases.


Asunto(s)
Envejecimiento , Eje Cerebro-Intestino , Disfunción Cognitiva , Microbioma Gastrointestinal , Sirtuina 2 , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Ratones , Envejecimiento/efectos de los fármacos , Sirtuina 2/metabolismo , Masculino , Eje Cerebro-Intestino/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Simulación del Acoplamiento Molecular , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...