Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
iScience ; 27(5): 109686, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38660396

RESUMO

Psilocybin, and its metabolite psilocin, induces psychedelic effects through activation of the 5-HT2A receptor. Psilocybin has been proposed as a treatment for depression and anxiety but sometimes induces anxiety in humans. An understanding of mechanisms underlying the anxiety response will help to better develop therapeutic prospects of psychedelics. In the current study, psilocybin induced an acute increase in anxiety in behavioral paradigms in mice. Importantly, pharmacological blocking of the 5-HT2A receptor attenuates psilocybin-induced head twitch response, a behavioral proxy for the psychedelic response, but does not rescue psilocybin's effect on anxiety-related behavior. Phosphopeptide analysis in the amygdala uncovered signal transduction pathways that are dependent or independent of the 5-HT2A receptor. Furthermore, presynaptic proteins are specifically involved in psilocybin-induced acute anxiety. These insights into how psilocybin may induce short-term anxiety are important for understanding how psilocybin may best be used in the clinical framework.

2.
NPJ Biofilms Microbiomes ; 9(1): 103, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38110423

RESUMO

Autism Spectrum Disorder (ASD) is a neurodevelopmental condition which is defined by decreased social communication and the presence of repetitive or stereotypic behaviors. Recent evidence has suggested that the gut-brain axis may be important in neurodevelopment in general and may play a role in ASD in particular. Here, we present a study of the gut microbiome in 96 individuals diagnosed with ASD in Israel, compared to 42 neurotypical individuals. We determined differences in alpha and beta diversity in the microbiome of individuals with ASD and demonstrated that the phylum Bacteroidetes and genus Bacteroides were the most significantly over-represented in individuals with ASD. To understand the possible functional significance of these changes, we treated newborn mice with Bacteroides fragilis at birth. B. fragilis-treated male mice displayed social behavior dysfunction, increased repetitive behaviors, and gene expression dysregulation in the prefrontal cortex, while female mice did not display behavioral deficits. These findings suggest that overabundance of Bacteroides, particularly in early life, may have functional consequences for individuals with ASD.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Humanos , Masculino , Camundongos , Feminino , Animais , Transtorno do Espectro Autista/diagnóstico , Transtorno do Espectro Autista/genética , Bacteroides/genética , Modelos Animais de Doenças , Comportamento Social
3.
Transl Psychiatry ; 13(1): 305, 2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37783686

RESUMO

Autism is a neurodevelopmental disorder characterized by early-onset social behavioral deficits and repetitive behaviors. Chromodomain helicase DNA-binding protein (CHD8) is among the genes most strongly associated with autism. In addition to the core behavioral symptoms of autism, affected individuals frequently present with gastrointestinal symptoms that are also common among individuals harboring mutations in the gene encoding CHD8. However, little is known regarding the mechanisms whereby CHD8 affects gut function. In addition, it remains unknown whether gastrointestinal manifestations contribute to the behavioral phenotypes of autism. The current study found that mice haploinsufficient for the large isoform of Chd8 (Chd8L) exhibited increased intestinal permeability, transcriptomic dysregulation in gut epithelial cells, reduced tuft cell and goblet cell counts in the gut, and an overall increase in microbial load. Gut epithelial cell-specific Chd8 haploinsufficiency was associated with increased anxiety-related behaviors together with a decrease in tuft cell numbers. Antibiotic treatment of Chd8L haploinsufficient mice attenuated social behavioral deficits. Together, these results suggest Chd8 as a key determinant of autism-related gastrointestinal deficits, while also laying the ground for future studies on the link between GI deficits and autism-related behaviors.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Camundongos , Animais , Transtorno Autístico/genética , Eixo Encéfalo-Intestino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Ligação a DNA/genética , Transtorno do Espectro Autista/genética , Células Epiteliais
4.
Nat Neurosci ; 26(7): 1208-1217, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37365313

RESUMO

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by heterogeneous cognitive, behavioral and communication impairments. Disruption of the gut-brain axis (GBA) has been implicated in ASD although with limited reproducibility across studies. In this study, we developed a Bayesian differential ranking algorithm to identify ASD-associated molecular and taxa profiles across 10 cross-sectional microbiome datasets and 15 other datasets, including dietary patterns, metabolomics, cytokine profiles and human brain gene expression profiles. We found a functional architecture along the GBA that correlates with heterogeneity of ASD phenotypes, and it is characterized by ASD-associated amino acid, carbohydrate and lipid profiles predominantly encoded by microbial species in the genera Prevotella, Bifidobacterium, Desulfovibrio and Bacteroides and correlates with brain gene expression changes, restrictive dietary patterns and pro-inflammatory cytokine profiles. The functional architecture revealed in age-matched and sex-matched cohorts is not present in sibling-matched cohorts. We also show a strong association between temporal changes in microbiome composition and ASD phenotypes. In summary, we propose a framework to leverage multi-omic datasets from well-defined cohorts and investigate how the GBA influences ASD.


Assuntos
Transtorno do Espectro Autista , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/genética , Eixo Encéfalo-Intestino , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Estudos Transversais , Teorema de Bayes , Reprodutibilidade dos Testes , Citocinas
5.
Int J Mol Sci ; 23(16)2022 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-36012331

RESUMO

The non-receptor focal adhesion kinase (FAK) is highly expressed in the central nervous system during development, where it regulates neurite outgrowth and axon guidance, but its role in the adult healthy and diseased brain, specifically in Alzheimer's disease (AD), is largely unknown. Using the 3xTg-AD mouse model, which carries three mutations associated with familial Alzheimer's disease (APP KM670/671NL Swedish, PSEN1 M146V, MAPT P301L) and develops age-related progressive neuropathology including amyloid plaques and Tau tangles, we describe here, for the first time, the in vivo role of FAK in AD pathology. Our data demonstrate that while site-specific knockdown in the hippocampi of 3xTg-AD mice has no effect on learning and memory, hippocampal overexpression of the protein leads to a significant decrease in learning and memory capabilities, which is accompanied by a significant increase in amyloid ß (Aß) load. Furthermore, neuronal morphology is altered following hippocampal overexpression of FAK in these mice. High-throughput proteomics analysis of total and phosphorylated proteins in the hippocampi of FAK overexpressing mice indicates that FAK controls AD-like phenotypes by inhibiting cytoskeletal remodeling in neurons which results in morphological changes, by increasing Tau hyperphosphorylation, and by blocking astrocyte differentiation. FAK activates cell cycle re-entry and consequent cell death while downregulating insulin signaling, thereby increasing insulin resistance and leading to oxidative stress. Our data provide an overview of the signaling networks by which FAK regulates AD pathology and identify FAK as a novel therapeutic target for treating AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Proteína-Tirosina Quinases de Adesão Focal , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Transtornos da Memória/metabolismo , Camundongos , Camundongos Transgênicos , Placa Amiloide/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
6.
Mol Brain ; 15(1): 30, 2022 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379308

RESUMO

CCCTC-binding factor (CTCF) is a regulator of chromatin organization and has direct effects on gene transcription. Mutations in CTCF have been identified in individuals with neurodevelopmental conditions. There are wide range of behaviors associated with these mutations, including intellectual disabilities, changes in temperament, and autism. Previous mice-model studies have identified roles for CTCF in excitatory neurons in specific behaviors, particularly in regards to learning and memory. However, the role of CTCF in inhibitory neurons is less well defined. In the current study, specific knockout of CTCF in parvalbumin-expressing neurons, a subset of inhibitory neurons, induced a specific behavioral phenotype, including locomotor abnormalities, anxiolytic behavior, and a decrease in social behavior. The anxiolytic and social abnormalities are detected before the onset of locomotor abnormalities. Immunohistochemical analysis revealed a disbalance in parvalbumin-expressing and somatostatin-expressing cells in these mice. Single nuclei RNA sequencing identified changes in gene expression in parvalbumin-expressing neurons that are specific to inhibitory neuronal identity and function. Electrophysiology analysis revealed an enhanced inhibitory tone in the hippocampal pyramidal neurons in knockout mice. These findings indicate that CTCF in parvalbumin-expressing neurons has a significant role in the overall phenotype of CTCF-associated neurodevelopmental deficits.


Assuntos
Neurônios , Parvalbuminas , Animais , Ansiedade , Fator de Ligação a CCCTC , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Parvalbuminas/metabolismo , Comportamento Social
7.
Biol Rev Camb Philos Soc ; 97(2): 582-599, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34734461

RESUMO

The microbiome influences the emotional and cognitive phenotype of its host, as well as the neurodevelopment and pathophysiology of various brain processes and disorders, via the well-established microbiome-gut-brain axis. Rapidly accumulating data link the microbiome to severe neuropsychiatric disorders in humans, including schizophrenia, Alzheimer's and Parkinson's. Moreover, preclinical work has shown that perturbation of the microbiome is closely associated with social, cognitive and behavioural deficits. The potential of the microbiome as a diagnostic and therapeutic tool is currently undercut by a lack of clear mechanistic understanding of the microbiome-gut-brain axis. This review establishes the hypothesis that the mechanism by which this influence is carried out is synaptic plasticity - long-term changes to the physical and functional neuronal structures that enable the brain to undertake learning, memory formation, emotional regulation and more. By examining the different constituents of the microbiome-gut-brain axis through the lens of synaptic plasticity, this review explores the diverse aspects by which the microbiome shapes the behaviour and mental wellbeing of the host. Key elements of this complex bi-directional relationship include neurotransmitters, neuronal electrophysiology, immune mediators that engage with both the central and enteric nervous systems and signalling cascades that trigger long-term potentiation of synapses. The importance of establishing mechanistic correlations along the microbiome-gut-brain axis cannot be overstated as they hold the potential for furthering current understanding regarding the vast fields of neuroscience and neuropsychiatry. This review strives to elucidate the promising theory of microbiome-driven synaptic plasticity in the hope of enlightening current researchers and inspiring future ones.


Assuntos
Microbioma Gastrointestinal , Microbiota , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiologia , Plasticidade Neuronal , Neurônios/fisiologia
8.
Genes (Basel) ; 12(8)2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34440307

RESUMO

Chromodomain-helicase-DNA-binding protein 8 (CHD8) has been identified as one of the genes with the strongest association with autism. The CHD8 protein is a transcriptional regulator that is expressed in nearly all cell types and has been implicated in multiple cellular processes, including cell cycle, cell adhesion, neuronal development, myelination, and synaptogenesis. Considering the central role of CHD8 in the genetics of autism, a deeper understanding of the physiological functions of CHD8 is important to understand the development of the autism phenotype and potential therapeutic targets. Different CHD8 mutant mouse models were developed to determine autism-like phenotypes and to fully understand their mechanisms. Here, we review the current knowledge on CHD8, with an emphasis on mechanistic lessons gained from animal models that have been studied.


Assuntos
Transtorno do Espectro Autista/fisiopatologia , Proteínas de Ligação a DNA/fisiologia , Transtornos do Neurodesenvolvimento/fisiopatologia , Fatores de Transcrição/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Modelos Animais de Doenças , Humanos , Camundongos , Fenótipo , Fatores de Transcrição/genética
9.
Mol Neurobiol ; 58(5): 2322-2341, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33417228

RESUMO

Alzheimer's disease (AD) is the most common cause of senile dementia and one of the greatest medical, social, and economic challenges. According to a dominant theory, amyloid-ß (Aß) peptide is a key AD pathogenic factor. Aß-soluble species interfere with synaptic functions, aggregate gradually, form plaques, and trigger neurodegeneration. The AD-associated pathology affects numerous systems, though the substantial loss of cholinergic neurons and α7 nicotinic receptors (α7AChR) is critical for the gradual cognitive decline. Aß binds to α7AChR under various experimental settings; nevertheless, the functional significance of this interaction is ambiguous. Whereas the capability of low Aß concentrations to activate α7AChR is functionally beneficial, extensive brain exposure to high Aß concentrations diminishes α7AChR activity, contributes to the cholinergic deficits that characterize AD. Aß and snake α-neurotoxins competitively bind to α7AChR. Accordingly, we designed a chemically modified α-cobratoxin (mToxin) to inhibit the interaction between Aß and α7AChR. Subsequently, we examined mToxin in a set of original in silico, in vitro, ex vivo experiments, and in a murine AD model. We report that mToxin reversibly inhibits α7AChR, though it attenuates Aß-induced synaptic transmission abnormalities, and upregulates pathways supporting long-term potentiation and reducing apoptosis. Remarkably, mToxin demonstrates no toxicity in brain slices and mice. Moreover, its chronic intracerebroventricular administration improves memory in AD-model animals. Our results point to unique mToxin neuroprotective properties, which might be tailored for the treatment of AD. Our methodology bridges the gaps in understanding Aß-α7AChR interaction and represents a promising direction for further investigations and clinical development.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Neurotoxinas/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Apoptose/efeitos dos fármacos , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/metabolismo , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Teóricos , Neurotoxinas/uso terapêutico , Ligação Proteica/efeitos dos fármacos
10.
Transl Psychiatry ; 10(1): 412, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33239620

RESUMO

The etiology of Autism Spectrum Disorders (ASD) includes a strong genetic component and a complicated environmental component. Recent evidence indicates that maternal diabetes, including gestational diabetes, is associated with an increased prevalence of ASD. While previous studies have looked into possible roles for maternal diabetes in neurodevelopment, there are few studies into how gestational diabetes, with no previous diabetic or metabolic phenotype, may affect neurodevelopment. In this study, we have specifically induced gestational diabetes in mice, followed by behavioral and molecular phenotyping of the mice offspring. Pregnant mice were injected with STZ a day after initiation of pregnancy. Glucose levels increased to diabetic levels between E7 and E14 in pregnancy in a subset of the pregnant animals. Male offspring of Gestational Diabetic mothers displayed increased repetitive behaviors with no dysregulation in the three-chambered social interaction test. RNA-seq analysis revealed a dysregulation in genes related to forebrain development in the frontal cortex and a dysregulation of a network of neurodevelopment and immune related genes in the striatum. Together, these results give evidence that gestational diabetes can induce changes in adulthood behavior and gene transcription in the brain.


Assuntos
Transtorno do Espectro Autista , Diabetes Gestacional , Efeitos Tardios da Exposição Pré-Natal , Animais , Cognição , Diabetes Gestacional/genética , Feminino , Lobo Frontal , Masculino , Camundongos , Gravidez , Transcrição Gênica
12.
Curr Opin Neurobiol ; 62: 76-82, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31972462

RESUMO

There is growing evidence for the involvement of the gut-microbiota in the regulation of emotions, behavior, and higher cognitive functions through the 'microbiome-gut-brain axis'. This relationship between the gut microbiota and the brain is pivotal for the development of the newborn, which receives its commensal microbiota at birth; dysbiosis may result in altered neurodevelopment. The hypothalamus-pituitary-adrenocortical (HPA) axis is actively involved in the stress response but is undeveloped in the newborn. Here, we describe how changes in the commensal microbiota influence the normal development of the HPA axis and review recent findings describing the essential crosstalk between the gut microbiota and the HPA axis and suggesting a role for the maternal and commensal microbiota in the development of the HPA axis and of the stress response.


Assuntos
Microbiota , Ansiedade , Encéfalo , Humanos , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Estresse Psicológico
13.
J Strength Cond Res ; 34(2): 422-429, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29927893

RESUMO

Martins, RS, Girouard, P, Elliott, E, and Mekary, S. Physiological responses of a jaw repositioning custom-made mouthguard on airway and their effects on athletic performance. J Strength Cond Res 34(2): 422-429, 2020-Advanced dental techniques such as jaw-repositioning have shown to increase lower body muscular power such as vertical jump, but its effects on acceleration and speed have not been studied. Similarly, jaw repositioning is commonly used to increase airways volume and ventilation in a special population (i.e., obstructive sleep apnea); however, its ergogenic effects on aerobic performance have yet not been studied. The purpose of the cross-over study was to investigate the effects of a jaw-repositioning custom-made mouthguard (JCM) on volumetric changes in airway and jaw position and determine the effects this may have on aerobic and anaerobic performance. Results indicated that jaw-repositioning custom-made mouthguard may have an ergogenic effect on performance. The JCM condition showed an increase of 13% in upper airway volume (p = 0.04), 10% in upper airway width (p = 0.004), 7% in ventilation (p = 0.006), 5% in maximal aerobic power (p = 0.003), 4% in time to exhaustion (p = 0.03), 3% in vertical jump (p = 0.03), 2% in broad jump (p = 0.009), and a decrease of 4% in 20-m (p = 0.04) and 2% in 40-m (p = 0.001) sprint times. This is the first study to demonstrate a significant link between jaw repositioning, airway volumetric change, and performance enhancement in both aerobic and anaerobic performances. The results of this study may lead to a change in culture for the use of mouthguards in different sports applications, from high orofacial injury risk sports to other sports, specifically for ergogenic enhancement.


Assuntos
Desempenho Atlético/fisiologia , Arcada Osseodentária/fisiologia , Protetores Bucais , Mecânica Respiratória/fisiologia , Estudos Cross-Over , Humanos , Masculino , Respiração , Testes de Função Respiratória , Adulto Jovem
14.
Nat Neurosci ; 22(11): 1913-1924, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31591560

RESUMO

Axonal injury results in regenerative success or failure, depending on whether the axon lies in the peripheral or the CNS, respectively. The present study addresses whether epigenetic signatures in dorsal root ganglia discriminate between regenerative and non-regenerative axonal injury. Chromatin immunoprecipitation for the histone 3 (H3) post-translational modifications H3K9ac, H3K27ac and H3K27me3; an assay for transposase-accessible chromatin; and RNA sequencing were performed in dorsal root ganglia after sciatic nerve or dorsal column axotomy. Distinct histone acetylation and chromatin accessibility signatures correlated with gene expression after peripheral, but not central, axonal injury. DNA-footprinting analyses revealed new transcriptional regulators associated with regenerative ability. Machine-learning algorithms inferred the direction of most of the gene expression changes. Neuronal conditional deletion of the chromatin remodeler CCCTC-binding factor impaired nerve regeneration, implicating chromatin organization in the regenerative competence. Altogether, the present study offers the first epigenomic map providing insight into the transcriptional response to injury and the differential regenerative ability of sensory neurons.


Assuntos
Axônios/fisiologia , Epigenômica , Gânglios Espinais/fisiologia , Regeneração Nervosa/fisiologia , Células Receptoras Sensoriais/fisiologia , Acetilação , Algoritmos , Animais , Fator de Ligação a CCCTC/genética , Cromatina/metabolismo , Feminino , Gânglios Espinais/lesões , Expressão Gênica , Histonas/metabolismo , Aprendizado de Máquina , Masculino , Camundongos , Camundongos Transgênicos , Nervo Isquiático/lesões , Análise de Sequência de RNA
15.
Transl Psychiatry ; 9(1): 257, 2019 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-31624228

RESUMO

Autism spectrum disorder (ASD) presents a wide, and often varied, behavioral phenotype. Improper assessment of risks has been reported among individuals diagnosed with ASD. Improper assessment of risks may lead to increased accidents and self-injury, also reported among individuals diagnosed with ASD. However, there is little knowledge of the molecular underpinnings of the impaired risk-assessment phenotype. In this study, we have identified impaired risk-assessment activity in multiple male ASD mouse models. By performing network-based analysis of striatal whole transcriptome data from each of these ASD models, we have identified a cluster of glutamate receptor-associated genes that correlate with the risk-assessment phenotype. Furthermore, pharmacological inhibition of striatal glutamatergic receptors was able to mimic the dysregulation in risk-assessment. Therefore, this study has identified a molecular mechanism that may underlie risk-assessment dysregulation in ASD.


Assuntos
Transtorno do Espectro Autista/genética , Corpo Estriado/metabolismo , Receptores de Glutamato/genética , Animais , Transtorno do Espectro Autista/etiologia , Modelos Animais de Doenças , Redes Reguladoras de Genes , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos , Proteínas do Tecido Nervoso/genética , Transcriptoma
16.
Front Behav Neurosci ; 13: 123, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31231198

RESUMO

During the past decade, there has been a substantial rise in the knowledge about the effects of gut microbiota on host physiology and behavior, including depressive behavior. Initial studies determined that gut microbiota can regulate host tryptophan levels, which is a main serotonin precursor. A dysfunctional serotonergic system is considered to be one of the main factors contributing to the development of depression. Therefore, we hypothesized that regulation of brain tryptophan and serotonin can explain, at least partly, the effects of microbiota on depressive behavior. To test this hypothesis, we examined depressive-like behavior and brain levels of serotonin and tryptophan, of germ free (GF) and specific-pathogen free (SPF) mice under basal conditions, or after acute tryptophan depletion (ATD) procedure, which is a method to decrease tryptophan and serotonin levels in the brain. In basal conditions, GF mice exhibited less depressive-like behavior in sucrose preference, tail-suspension and forced swim tests, compared to SPF mice. In addition, in mice that were not subjected to ATD, GF mice displayed higher levels of tryptophan, serotonin and 5-hydroxyindoleacetic acid (the main degradation product of serotonin) in medial prefrontal cortex (mPFC) and hippocampus (HIPPO), compared to SPF mice. Interestingly, ATD increased depressive-like behavior of GF, but not of SPF mice. These behavioral changes were accompanied by a stronger reduction of tryptophan, serotonin and 5-hydroxyindoleacetic acid in mPFC and HIPPO in GF mice after ATD, when compared to SPF mice. Therefore, the serotonergic system of GF mice is more vulnerable to the acute challenge of tryptophan reduction, and GF mice after tryptophan reduction behave more similarly to SPF mice. These data provide functional evidence that microbiota affects depression-like behavior through influencing brain tryptophan accessibility and the serotonergic system.

17.
Transl Psychiatry ; 9(1): 133, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30967529

RESUMO

Accumulating evidence demonstrates that the gut microbiota affects brain function and behavior, including depressive behavior. Antidepressants are the main drugs used for treatment of depression. We hypothesized that antidepressant treatment could modify gut microbiota which can partially mediate their antidepressant effects. Mice were chronically treated with one of five antidepressants (fluoxetine, escitalopram, venlafaxine, duloxetine or desipramine), and gut microbiota was analyzed, using 16s rRNA gene sequencing. After characterization of differences in the microbiota, chosen bacterial species were supplemented to vehicle and antidepressant-treated mice, and depressive-like behavior was assessed to determine bacterial effects. RNA-seq analysis was performed to determine effects of bacterial treatment in the brain. Antidepressants reduced richness and increased beta diversity of gut bacteria, compared to controls. At the genus level, antidepressants reduced abundances of Ruminococcus, Adlercreutzia, and an unclassified Alphaproteobacteria. To examine implications of the dysregulated bacteria, we chose one of antidepressants (duloxetine) and investigated if its antidepressive effects can be attenuated by simultaneous treatment with Ruminococcus flavefaciens or Adlercreutzia equolifaciens. Supplementation with R. flavefaciens diminished duloxetine-induced decrease in depressive-like behavior, while A. equolifaciens had no such effect. R. flavefaciens treatment induced changes in cortical gene expression, up-regulating genes involved in mitochondrial oxidative phosphorylation, while down-regulating genes involved in neuronal plasticity. Our results demonstrate that various types of antidepressants alter gut microbiota composition, and further implicate a role for R. flavefaciens in alleviating depressive-like behavior. Moreover, R. flavefaciens affects gene networks in the brain, suggesting a mechanism for microbial regulation of antidepressant treatment efficiency.


Assuntos
Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Depressão/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Ruminococcus/fisiologia , Animais , Comportamento Animal , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Ribossômico 16S/genética , Ruminococcus/efeitos dos fármacos
18.
Neurotherapeutics ; 15(4): 1036-1054, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30288668

RESUMO

The urea cycle is strongly implicated in the pathogenesis of Alzheimer's disease (AD). Arginase-I (ARGI) accumulation at sites of amyloid-beta (Aß) deposition is associated with L-arginine deprivation and neurodegeneration. An interaction between the arginase II (ARGII) and mTOR-ribosomal protein S6 kinase ß-1 (S6K1) pathways promotes inflammation and oxidative stress. In this study, we treated triple-transgenic (3×Tg) mice exhibiting increased S6K1 activity and wild-type (WT) mice with L-norvaline, which inhibits both arginase and S6K1. The acquisition of spatial memory was significantly improved in the treated 3×Tg mice, and the improvement was associated with a substantial reduction in microgliosis. In these mice, increases in the density of dendritic spines and expression levels of neuroplasticity-related proteins were followed by a decline in the levels of Aß toxic oligomeric and fibrillar species in the hippocampus. The findings point to an association of local Aß-driven and immune-mediated responses with altered L-arginine metabolism, and they suggest that arginase and S6K1 inhibition by L-norvaline may delay the progression of AD.


Assuntos
Doença de Alzheimer/complicações , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Valina/análogos & derivados , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Arginase/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/ultraestrutura , Transtornos Cognitivos/patologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Espinhas Dendríticas/ultraestrutura , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Microglia/patologia , Mutação/genética , Neurônios/efeitos dos fármacos , Neurônios/patologia , Presenilina-1/genética , Valina/uso terapêutico , Proteínas tau/genética
19.
Transl Neurosci ; 9: 43-53, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29876138

RESUMO

Arginine is one of the most versatile semi-essential amino acids. Further to the primary role in protein biosynthesis, arginine is involved in the urea cycle, and it is a precursor of nitric oxide. Arginine deficiency is associated with neurodegenerative diseases such as Parkinson's, Huntington's and Alzheimer's diseases (AD). In this study, we administer arginine intracerebroventricularly in a murine model of AD and evaluate cognitive functions in a set of behavioral tests. In addition, the effect of arginine on synaptic plasticity was tested electrophysiologically by assessment of the hippocampal long-term potentiation (LTP). The effect of arginine on ß amyloidosis was tested immunohistochemically. A role of arginine in the prevention of cytotoxicity and apoptosis was evaluated in vitro on PC-12 cells. The results indicate that intracerebroventricular administration of arginine improves spatial memory acquisition in 3xTg-AD mice, however, without significantly reducing intraneuronal ß amyloidosis. Arginine shows little or no impact on LTP and does not rescue LTP deterioration induced by Aß. Nevertheless, arginine possesses neuroprotective and antiapoptotic properties.

20.
Brain Behav Immun ; 73: 310-319, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29787855

RESUMO

Recent studies have determined that the microbiome has direct effects on behavior, and may be dysregulated in neurodevelopmental conditions. Considering that neurodevelopmental conditions, such as autism, have a strong genetic etiology, it is necessary to understand if genes associated with neurodevelopmental disorders, such as Shank3, can influence the gut microbiome, and if probiotics can be a therapeutic tool. In this study, we have identified dysregulation of several genera and species of bacteria in the gut and colon of both male and female Shank3 KO mice. L. reuteri, a species with decreased relative abundance in the Shank3 KO mice, positively correlated with the expression of gamma-Aminobutyric acid (GABA) receptor subunits in the brain. Treatment of Shank3 KO mice with L. reuteri induced an attenuation of unsocial behavior specifically in male Shank3 mice, and a decrease in repetitive behaviors in both male and female Shank3 KO mice. In addition, L. reuteri treatment affected GABA receptor gene expression and protein levels in multiple brain regions. This study identifies bacterial species that are sensitive to an autism-related mutation, and further suggests a therapeutic potential for probiotic treatment.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/microbiologia , Microbioma Gastrointestinal/genética , Animais , Transtorno do Espectro Autista/metabolismo , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Modelos Animais de Doenças , Disbiose/microbiologia , Feminino , Microbioma Gastrointestinal/fisiologia , Limosilactobacillus reuteri/genética , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos , Modelos Genéticos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Probióticos/metabolismo , Probióticos/farmacologia , Probióticos/uso terapêutico , Receptores de GABA/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA