Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Psychiatry Neurosci ; 49(1): E35-E44, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38302137

RESUMO

BACKGROUND: Susceptibility to schizophrenia is determined by interactions between genes and environment, possibly via epigenetic mechanisms. Schizophrenia has been associated with a restrictive epigenome, and histone deacetylase (HDAC) inhibitors have been postulated as coadjuvant agents to potentiate the efficacy of current antipsychotic drugs. We aimed to evaluate global histone posttranslational modifications (HPTMs) and HDAC expression and activity in the dorsolateral prefrontal cortex (DLPFC) of individuals with schizophrenia. METHODS: We used postmortem DLPFC samples of individuals with schizophrenia and controls matched for sex, age, and postmortem interval. Schizophrenia samples were classified into antipsychotic-treated or antipsychotic-free subgroups according to blood toxicology. Expression of HPTMs and HDAC was quantified by Western blot. HDAC activity was measured with a fluorometric assay. RESULTS: H3K9ac, H3K27ac, and H3K4me3 were globally enhanced in the DLPFC of individuals with schizophrenia (+24%-42%, p < 0.05). HDAC activity (-17%, p < 0.01) and HDAC4 protein expression (-20%, p < 0.05) were downregulated in individuals with schizophrenia. Analyses of antipsychotic-free and antipsychotic-treated subgroups revealed enhanced H3K4me3 and H3K27ac (+24%-49%, p < 0.05) and reduced HDAC activity in the antipsychotic-treated, but not in the antipsychotic-free subgroup. LIMITATIONS: Special care was taken to control the effect of confounding factors: age, sex, postmortem interval, and storage time. However, replication studies in bigger cohorts might strengthen the association between permissive HPTMs and schizophrenia. CONCLUSION: We found global HPTM alterations consistent with an aberrantly permissive epigenome in schizophrenia. Further studies to elucidate the significance of enhanced permissive HPTMs in schizophrenia and its association with the mechanism of action of antipsychotic drugs are encouraged.


Assuntos
Antipsicóticos , Esquizofrenia , Humanos , Esquizofrenia/tratamento farmacológico , Esquizofrenia/genética , Esquizofrenia/metabolismo , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Córtex Pré-Frontal Dorsolateral , Histonas , Epigênese Genética , Córtex Pré-Frontal/metabolismo
2.
Int J Mol Sci ; 25(13)2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-39000260

RESUMO

Extracellular vesicles (EVs) constitute a sophisticated molecular exchange mechanism highly regarded for their potential as a next-generation platform for compound delivery. However, identifying sustainable and biologically safe sources of EVs remains a challenge. This work explores the emergence of novel sources of plant and bacterial-based EVs, such as those obtained from food industry by-products, known as BP-EVs, and their potential to be used as safer and biocompatible nanocarriers, addressing some of the current challenges of the field. These novel sources exhibit remarkable oral bioavailability and biodistribution, with minimal cytotoxicity and a selective targeting capacity toward the central nervous system, liver, and skeletal tissues. Additionally, we review the ease of editing these recently uncovered nanocarrier-oriented vesicles using common EV editing methods, examining the cargo-loading processes applicable to these sources, which involve both passive and active functionalization methods. While the primary focus of these novel sources of endogenous EVs is on molecule delivery to the central nervous system and skeletal tissue based on their systemic target preference, their use, as reviewed here, extends beyond these key applications within the biotechnological and biomedical fields.


Assuntos
Bactérias , Sistemas de Liberação de Medicamentos , Vesículas Extracelulares , Plantas , Vesículas Extracelulares/metabolismo , Bactérias/metabolismo , Humanos , Plantas/metabolismo , Animais , Sistemas de Liberação de Medicamentos/métodos , Leveduras/metabolismo , Portadores de Fármacos/química
3.
Int J Mol Sci ; 24(3)2023 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-36768626

RESUMO

The crosstalk between the opioidergic system and mitogen-activated protein kinases (MAPKs) has a critical role in mediating stress-induced behaviors related to the pathophysiology of anxiety. The present study evaluated the basal status and stress-induced alterations of cortico-thalamic MAPKs and other cell fate-related signaling pathways potentially underlying the anxiogenic endophenotype of PDYN gene-deficient mice. Compared to littermates, PDYN knockout (KO) mice had lower cortical and or thalamic amounts of the phospho-activated MAPKs c-Jun N-terminal kinase (JNK1/2) and extracellular signal-regulated kinase (ERK1/2). Similarly, PDYN-KO animals displayed reduced cortico-thalamic densities of total and phosphorylated (at Ser191) species of the cell fate regulator Fas-associated protein with death domain (FADD) without alterations in the Fas receptor. Exposure to acute restraint and chronic mild stress stimuli induced the robust stimulation of JNK1/2 and ERK1/2 MAPKs, FADD, and Akt-mTOR pathways, without apparent increases in apoptotic rates. Interestingly, PDYN deficiency prevented stress-induced JNK1/2 and FADD but not ERK1/2 or Akt-mTOR hyperactivations. These findings suggest that cortico-thalamic MAPK- and FADD-dependent neuroplasticity might be altered in PDYN-KO mice. In addition, the results also indicate that the PDYN gene (and hence dynorphin release) may be required to stimulate JNK1/2 and FADD (but not ERK1/2 or Akt/mTOR) pathways under environmental stress conditions.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Apoptose/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Alzheimers Dement ; 18(11): 2023-2035, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-34994517

RESUMO

INTRODUCTION: Physical activity (PA) is widely recommended for age-related brain health, yet its neurobiology is not well understood. Animal models indicate PA is synaptogenic. We examined the relationship between PA and synaptic integrity markers in older adults. METHODS: Four hundred four decedents from the Rush Memory and Aging Project completed annual actigraphy monitoring (Mean visits = 3.5±2.4) and post mortem evaluation. Brain tissue was analyzed for presynaptic proteins (synaptophysin, synaptotagmin-1, vesicle-associated membrane proteins, syntaxin, complexin-I, and complexin-II), and neuropathology. Models examined relationships between late-life PA (averaged across visits), and timing-specific PA (time to autopsy) with synaptic proteins. RESULTS: Greater late-life PA associated with higher presynaptic protein levels (0.14 < ß < 0.20), except complexin-II (ß = 0.08). Relationships were independent of pathology but timing specific; participants who completed actigraphy within 2 years of brain tissue measurements showed largest PA-to-synaptic protein associations (0.32 < ß < 0.38). Relationships between PA and presynaptic proteins were comparable across brain regions sampled. DISCUSSION: PA associates with synaptic integrity in a regionally global, but time-linked nature in older adults.


Assuntos
Encéfalo , Exercício Físico , Animais , Envelhecimento/patologia , Actigrafia
5.
Acta Neuropathol ; 141(5): 755-770, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33646358

RESUMO

Age-related neuropathologies progressively impair cognitive abilities by damaging synaptic function. We aimed to identify key components within the presynaptic SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) machinery associated with cognitive performance and estimate their potential contribution to brain reserve in old age. We used targeted SRM proteomics to quantify amounts of 60 peptides, encoded in 30 different genes, in postmortem specimens of the prefrontal cortex from 1209 participants of two aging studies, with available antemortem cognitive evaluations and postmortem neuropathologic assessments. We found that select (but not all) proteoforms are strongly associated with cognitive function and the burden of Alzheimer's disease (AD) pathology. Specifically, greater abundance of STX1A (but not other syntaxins), SYT12, full-length SNAP25, and the GABAergic STXBP1 variant were robustly associated with better cognitive performance. By contrast, greater abundance of other presynaptic proteins (e.g., STXBP5 or tomosyn, STX7, or SYN2) showed a negative influence on cognition. Regression models adjusting for demographic and pathologic variables showed that altered levels of these protein species explained 7.7% additional between-subject variance in cognition (more than any individual age-related neuropathology in the model), suggesting that these molecules constitute key elements of brain reserve. Network analyses indicated that those peptides associated with brain reserve, and closest to the SNARE fusogenic activity, showed greater centrality measures and were better connected in the network. Validation assays confirmed the selective loss of the STX1A (but not STX1B) isoform in cognitively impaired cases. In rodent and human brains, STX1A was selectively located at glutamatergic terminals. However, in AD brains, STX1A was redistributed adjacent to neuritic pathology, and markedly expressed in astrocytes. Our study provides strong evidence, indicating that select presynaptic proteins are key in maintaining brain reserve. Compromised ability to sustain expression levels of these proteins may trigger synaptic dysfunction and concomitant cognitive impairment.


Assuntos
Encéfalo/metabolismo , Cognição/fisiologia , Reserva Cognitiva/fisiologia , Proteínas SNARE/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/patologia , Feminino , Humanos , Masculino , Proteômica , Ratos , Ratos Sprague-Dawley
6.
Neurobiol Dis ; 114: 31-44, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29496544

RESUMO

The molecular underpinnings associated with cognitive reserve remain poorly understood. Because animal models fail to fully recapitulate the complexity of human brain aging, postmortem studies from well-designed cohorts are crucial to unmask mechanisms conferring cognitive resistance against cumulative neuropathologies. We tested the hypothesis that functionality of the SNARE protein interactome might be an important resilience factor preserving cognitive abilities in old age. Cognition was assessed annually in participants from the Rush "Memory and Aging Project" (MAP), a community-dwelling cohort representative of the overall aging population. Associations between cognition and postmortem neurochemical data were evaluated in functional assays quantifying various species of the SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) machinery in samples from the inferior temporal (IT, n = 154) and middle-frontal (MF, n = 174) gyri. Using blue-native gel electrophoresis, we isolated and quantified several types of complexes containing the three SNARE proteins (syntaxin-1, SNAP25, VAMP), as well as the GABAergic/glutamatergic selectively expressed complexins-I/II (CPLX1/2), in brain tissue homogenates and reconstitution assays with recombinant proteins. Multivariate analyses revealed significant associations between IT and MF neurochemical data (SNARE proteins and/or complexes), and multiple age-related neuropathologies, as well as with multiple cognitive domains of MAP participants. Controlling for demographic variables, neuropathologic indices and total synapse density, we found that temporal 150-kDa SNARE species (representative of pan-synaptic functionality) and frontal CPLX1/CPLX2 ratio of 500-kDa heteromeric species (representative of inhibitory/excitatory input functionality) were, among all the immunocharacterized complexes, the strongest predictors of cognitive function nearest death. Interestingly, these two neurochemical variables were associated with different cognitive domains. In addition, linear mixed effect models of global cognitive decline estimated that both 150-kDa SNARE levels and CPLX1/CPLX2 ratio were associated with better cognition and less decline over time. The results are consistent with previous studies reporting that synapse dysfunction (i.e. dysplasticity) may be initiated early, and relatively independent of neuropathology-driven synapse loss. Frontotemporal dysregulation of the GABAergic/glutamatergic stimuli might be a target for future drug development.


Assuntos
Envelhecimento/fisiologia , Disfunção Cognitiva/fisiopatologia , Lobo Frontal/fisiopatologia , Proteínas SNARE/fisiologia , Lobo Temporal/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/genética , Envelhecimento/patologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/patologia , Feminino , Lobo Frontal/patologia , Humanos , Masculino , Lobo Temporal/patologia
7.
PLoS Med ; 14(4): e1002287, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28441426

RESUMO

INTRODUCTION: The molecular underpinnings of the dissociation of cognitive performance and neuropathological burden are poorly understood, and there are currently no known genetic or epigenetic determinants of the dissociation. METHODS AND FINDINGS: "Residual cognition" was quantified by regressing out the effects of cerebral pathologies and demographic characteristics on global cognitive performance proximate to death. To identify genes influencing residual cognition, we leveraged neuropathological, genetic, epigenetic, and transcriptional data available for deceased participants of the Religious Orders Study (n = 492) and the Rush Memory and Aging Project (n = 487). Given that our sample size was underpowered to detect genome-wide significance, we applied a multistep approach to identify genes influencing residual cognition, based on our prior observation that independent genetic and epigenetic risk factors can converge on the same locus. In the first step (n = 979), we performed a genome-wide association study with a predefined suggestive p < 10-5, and nine independent loci met this threshold in eight distinct chromosomal regions. Three of the six genes within 100 kb of the lead SNP are expressed in the dorsolateral prefrontal cortex (DLPFC): UNC5C, ENC1, and TMEM106B. In the second step, in the subset of participants with DLPFC DNA methylation data (n = 648), we found that residual cognition was related to differential DNA methylation of UNC5C and ENC1 (false discovery rate < 0.05). In the third step, in the subset of participants with DLPFC RNA sequencing data (n = 469), brain transcription levels of UNC5C and ENC1 were evaluated for their association with residual cognition: RNA levels of both UNC5C (estimated effect = -0.40, 95% CI -0.69 to -0.10, p = 0.0089) and ENC1 (estimated effect = 0.0064, 95% CI 0.0033 to 0.0096, p = 5.7 × 10-5) were associated with residual cognition. In secondary analyses, we explored the mechanism of these associations and found that ENC1 may be related to the previously documented effect of depression on cognitive decline, while UNC5C may alter the composition of presynaptic terminals. Of note, the TMEM106B allele identified in the first step as being associated with better residual cognition is in strong linkage disequilibrium with rs1990622A (r2 = 0.66), a previously identified protective allele for TDP-43 proteinopathy. Limitations include the small sample size for the genetic analysis, which was underpowered to detect genome-wide significance, the evaluation being limited to a single cortical region for epigenetic and transcriptomic data, and the use of categorical measures for certain non-amyloid-plaque, non-neurofibrillary-tangle neuropathologies. CONCLUSIONS: Through a multistep analysis of cognitive, neuropathological, genomic, epigenomic, and transcriptomic data, we identified ENC1 and UNC5C as genes with convergent genetic, epigenetic, and transcriptomic evidence supporting a potential role in the dissociation of cognition and neuropathology in an aging population, and we expanded our understanding of the TMEM106B haplotype that is protective against TDP-43 proteinopathy.


Assuntos
Envelhecimento/fisiologia , Encéfalo/patologia , Transtornos Cognitivos/genética , Cognição/fisiologia , Proteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/genética , Proteínas Nucleares/genética , Receptores de Superfície Celular/genética , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/genética , Alelos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Transtornos Cognitivos/metabolismo , Metilação de DNA , Depressão/metabolismo , Epigênese Genética , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Desequilíbrio de Ligação , Masculino , Proteínas de Membrana/metabolismo , Memória , Proteínas dos Microfilamentos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Netrina , Neuropeptídeos/metabolismo , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleotídeo Único , RNA/metabolismo , Receptores de Superfície Celular/metabolismo , Proteinopatias TDP-43/genética
8.
Acta Neuropathol ; 133(3): 395-407, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27866231

RESUMO

Progressive accumulation of Alzheimer's disease-related pathology is associated with cognitive dysfunction. Differences in cognitive reserve may contribute to individual differences in cognitive function in the presence of comparable neuropathology. The protective effects of cognitive reserve could contribute differentially in early versus late stages of the disease. We investigated presynaptic proteins as measures of brain reserve (a subset of total cognitive reserve), and used Braak staging to estimate the progression of Alzheimer's disease. Antemortem evaluations of cognitive function, postmortem assessments of pathologic indices, and presynaptic protein analyses, including the complexins I and II as respective measures of inhibitory and excitatory terminal function, were assayed in multiple key brain regions in 418 deceased participants from a community study. After covarying for demographic variables, pathologic indices, and overall synapse density, lower brain complexin-I and -II levels contributed to cognitive dysfunction (P < 0.01). Each complexin appeared to be dysregulated at a different Braak stage. Inhibitory complexin-I explained 14.4% of the variance in global cognition in Braak 0-II, while excitatory complexin-II explained 7.3% of the variance in Braak V-VI. Unlike other presynaptic proteins, complexins did not colocalize with pathologic tau within neuritic plaques, suggesting that these functional components of the synaptic machinery are cleared early from dystrophic neurites. Moreover, complexin levels showed distinct patterns of change related to memory challenges in a rat model, supporting the functional specificity of these proteins. The present results suggest that disruption of inhibitory synaptic terminals may trigger early cognitive impairment, while excitatory terminal disruption may contribute relatively more to later cognitive impairment.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Doença de Alzheimer/complicações , Encéfalo/metabolismo , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/patologia , Proteínas do Tecido Nervoso/metabolismo , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Animais , Autopsia , Encéfalo/patologia , Transtornos Cognitivos/metabolismo , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Masculino , Aprendizagem em Labirinto , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Long-Evans , Características de Residência , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
9.
Transl Psychiatry ; 14(1): 113, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38396013

RESUMO

Antipsychotic-induced low availability of group II metabotropic glutamate receptors (including mGlu2R and mGlu3R) in brains of schizophrenia patients may explain the limited efficacy of mGlu2/3R ligands in clinical trials. Studies evaluating mGlu2/3R levels in well-designed, large postmortem brain cohorts are needed to address this issue. Postmortem samples from the dorsolateral prefrontal cortex of 96 schizophrenia subjects and matched controls were collected. Toxicological analyses identified cases who were (AP+) or were not (AP-) receiving antipsychotic treatment near the time of death. Protein and mRNA levels of mGlu2R and mGlu3R, as well as GRM2 and GRM3 promoter-attached histone posttranslational modifications, were quantified. Experimental animal models were used to compare with data obtained in human tissues. Compared to matched controls, schizophrenia cortical samples had lower mGlu2R protein amounts, regardless of antipsychotic medication. Downregulation of mGlu3R was observed in AP- schizophrenia subjects only. Greater predicted occupancy values of dopamine D2 and serotonin 5HT2A receptors correlated with higher density of mGlu3R, but not mGlu2R. Clozapine treatment and maternal immune activation in rodents mimicked the mGlu2R, but not mGlu3R regulation observed in schizophrenia brains. mGlu2R and mGlu3R mRNA levels, and the epigenetic control mechanisms did not parallel the alterations at the protein level, and in some groups correlated inversely. Insufficient cortical availability of mGlu2R and mGlu3R may be associated with schizophrenia. Antipsychotic treatment may normalize mGlu3R, but not mGlu2R protein levels. A model in which epigenetic feedback mechanisms controlling mGlu3R expression are activated to counterbalance mGluR loss of function is described.


Assuntos
Antipsicóticos , Receptores de Glutamato Metabotrópico , Esquizofrenia , Animais , Humanos , Antipsicóticos/farmacologia , Antipsicóticos/uso terapêutico , Esquizofrenia/tratamento farmacológico , Esquizofrenia/genética , Esquizofrenia/metabolismo , Receptores de Glutamato Metabotrópico/genética , Encéfalo/metabolismo , Epigênese Genética , RNA Mensageiro/metabolismo
10.
Biomedicines ; 12(1)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38255234

RESUMO

Extracellular vesicles (EVs) are tiny membranous structures that mediate intercellular communication. The role(s) of these vesicles have been widely investigated in the context of neurological diseases; however, their potential implications in the neuropathology subjacent to human psychiatric disorders remain mostly unknown. Here, by using next-generation discovery-driven proteomics, we investigate the potential role(s) of brain EVs (bEVs) in schizophrenia (SZ) by analyzing these vesicles from the three post-mortem anatomical brain regions: the prefrontal cortex (PFC), hippocampus (HC), and caudate (CAU). The results obtained indicate that bEVs from SZ-affected brains contain region-specific proteins that are associated with abnormal GABAergic and glutamatergic transmission. Similarly, these vesicles from the analyzed regions were implicated in synaptic decay, abnormal brain immunity, neuron structural imbalances, and impaired cell homeostasis. Our findings also provide evidence, for the first time, that networks of molecular exchange (involving the PFC, HC, and CAU) are potentially active and mediated by EVs in non-diseased brains. Additionally, these bEV-mediated networks seem to have become partially reversed and largely disrupted in the brains of subjects affected by SZ. Taken as a whole, these results open the door to the uncovering of new biological markers and therapeutic targets, based on the compositions of bEVs, for the benefit of patients affected by SZ and related psychotic disorders.

11.
Int J Neuropsychopharmacol ; 16(3): 683-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22964075

RESUMO

Cyclin-dependent kinase-5 (CDK5) and p35/p25 activators, interacting with the exocytotic machinery (e.g. munc18-1 and syntaxin-1A), play critical roles in neurosecretion. The basal status of CDK5/p35/p25 and the effect of psychotropic drugs (detected in blood/urine samples) were investigated in post-mortem prefrontal cortex (PFC)/Brodmann's area 9 of schizophrenia (SZ) and major depression (MD) subjects. In SZ (all subjects, n = 24), CDK5 and p35, but not p25, were reduced (-28 to -58%) compared to controls. In SZ antipsychotic-free (n = 12), activator p35 was decreased (-52%). In SZ antipsychotic-treated (n = 12), marked reductions of CDK5 (-47%), p35 (-76%) and p25 (-36%) were quantified. In MD (n = 13), including antidepressant-free/treated subgroups, CDK5, p35 and p25 were unaltered. In SZ (n = 24), CDK5, p35 or p25 correlated with munc18-1a, but not with syntaxin-1A. The results demonstrate reduced p35 basal content and down-regulation of CDK5/p35/p25 by antipsychotics in SZ. The suggested CDK5/munc18-1a functional interaction may lead to dysregulated neurosecretion in SZ PFC.


Assuntos
Quinase 5 Dependente de Ciclina/fisiologia , Transtorno Depressivo Maior/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Córtex Pré-Frontal/metabolismo , Psicotrópicos/uso terapêutico , Esquizofrenia/metabolismo , Adulto , Idoso , Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Quinase 5 Dependente de Ciclina/biossíntese , Transtorno Depressivo Maior/patologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/biossíntese , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/patologia , Psicotrópicos/farmacologia , Estudos Retrospectivos , Esquizofrenia/tratamento farmacológico , Esquizofrenia/patologia
12.
Addict Biol ; 18(4): 633-43, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22741574

RESUMO

The treatment of cocaine addiction remains a challenge. The dopamine replacement approach in cocaine addiction involves the use of a competing dopaminergic agonist that might suppress withdrawal and drug craving in abstinent individuals. Although it has long been postulated that such an approach may be therapeutically successful, preclinical or clinical evidence showing its effectiveness to prevent relapse is scant. We used in rats a procedure that involved substitution of the N-substituted benztropine analog 3α-[bis(4'-fluorophenyl)methoxy]-tropane (AHN-1055), a long-acting dopamine uptake inhibitor (DUI), for cocaine. Maintenance treatment was self-administered. After extinction, reinstatement of drug seeking was induced by cocaine priming. We measured the contents of brain-derived neurotrophic factor (BDNF), c-Fos and Fas-associated death domain (FADD) proteins in the medial prefrontal cortex (mPFC) following reinstatement. DUI, but not amphetamine, substitution led to extinction of active lever presses, as did saline substitution. DUI substitution significantly reduced cocaine-induced reinstatement of drug-seeking behavior, which was strongly elicited after saline substitution. Rats passively yoked to DUI also showed reduced cocaine-primed reinstatement. Reductions in drug seeking during reinstatement were matched by downward shifts in the contents of BDNF, c-Fos and FADD proteins in the mPFC, which were elevated in relapsing rats. These data indicate that DUI substitution not only leads to extinction of self-administration behavior but also prevents reinstatement of drug seeking induced by cocaine re-exposure. Thus, DUI substitution therapy using compounds with low abuse potential, even if received passively in the context previously paired with drug taking, may provide an effective treatment for stimulant addiction.


Assuntos
Benzotropina/análogos & derivados , Transtornos Relacionados ao Uso de Cocaína/tratamento farmacológico , Cocaína/administração & dosagem , Inibidores da Captação de Dopamina/uso terapêutico , Comportamento de Procura de Droga/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Análise de Variância , Animais , Comportamento Aditivo/tratamento farmacológico , Comportamento Aditivo/metabolismo , Benzotropina/administração & dosagem , Benzotropina/farmacologia , Benzotropina/uso terapêutico , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cocaína/farmacologia , Transtornos Relacionados ao Uso de Cocaína/metabolismo , Inibidores da Captação de Dopamina/administração & dosagem , Inibidores da Captação de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Extinção Psicológica/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Long-Evans , Esquema de Reforço , Prevenção Secundária , Autoadministração , Síndrome de Abstinência a Substâncias/tratamento farmacológico
13.
Int J Neuropsychopharmacol ; 15(5): 573-88, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21669024

RESUMO

Munc18-1 and syntaxin-1 are crucial interacting molecules for synaptic membrane fusion and neurotransmitter release. Contrasting abnormalities of several proteins of the exocytotic machinery, including the formation of SNARE (synaptobrevin, SNAP-25 and syntaxin-1) complexes, have been reported in schizophrenia. This study quantified in the dorsolateral prefrontal cortex (PFC, Brodmann area 9) the immunocontent of munc18-1a/b isoforms, syntaxin-1A, other presynaptic proteins (synaptotagmin, synaptophysin), and SNARE complexes, as well as the effects of psychoactive drug exposure, in schizophrenia (SZ, n=24), non-schizophrenia suicide (SD, n=13) and major depression (MD, n=15) subjects compared to matched controls (n=39). SZ was associated with normal expression of munc18-1a/b and increased syntaxin-1A (+44%). The presence of antipsychotic drugs reduced the basal content of munc18-1a isoform (-23%) and synaptobrevin (-32%), and modestly reduced that of up-regulated syntaxin-1A (-16%). Munc18-1a and syntaxin-1A protein expression correlated positively in controls but showed a markedly opposite pattern in SZ, regardless of antipsychotic treatment. Thus, the ratio of syntaxin-1A to munc18-1a showed a net increase in SZ (+53/114%). The SNARE complex (75 kDa) was found unaltered in antipsychotic-free and reduced (-28%) in antipsychotic-treated SZ subjects. None of these abnormalities were observed in SD and MD subjects, unexposed or exposed to psychoactive drugs. The results reveal some exocytotic dysfunctions in SZ that are probably related to an imbalance of the interaction between munc18-1a and SNARE (mainly syntaxin-1A) complex. Moreover, antipsychotic drug treatment is associated with lower content of key proteins of the exocytotic machinery, which could result in a destabilization/impairment of neurosecretion.


Assuntos
Antipsicóticos/metabolismo , Transtorno Depressivo Maior/metabolismo , Proteínas Munc18/metabolismo , Córtex Pré-Frontal/metabolismo , Proteínas SNARE/metabolismo , Esquizofrenia , Suicídio , Sintaxina 1/metabolismo , Animais , Regulação para Baixo , Exocitose/fisiologia , Humanos , Masculino , Isoformas de Proteínas/metabolismo , Proteínas R-SNARE/metabolismo , Ratos , Ratos Sprague-Dawley , Esquizofrenia/tratamento farmacológico , Esquizofrenia/metabolismo , Sinaptotagminas/metabolismo
14.
Pharmacol Rep ; 73(4): 1122-1135, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33641090

RESUMO

BACKGROUND: Alpha2A-adrenergic (α2A-AR) and cannabinoid CB1 (CB1-R) receptors exert their functions modulating multiple signaling pathways, including MEK-ERK (extracellular signal-regulated kinases) and FADD (Fas-associated protein with death domain) cascades. These molecules are relevant in finding biased agonists with fewer side effects, but the mechanisms involving their modulations by α2A-AR- and CB1-R in vivo are unclear. This study investigated the roles of Gαi2 and Gαz proteins in mediating α2A-AR- and CB1-R-induced alterations of MEK-ERK and FADD phosphorylation (p-) in mouse brain cortex. METHODS: Gαi2 or Gαz protein knockdown was induced in mice with selective antisense oligodeoxinucleotides (ODNs; 3 nmol/day, 5 days) prior to UK-14,304 (UK or brimonidine; 1 mg/kg) or WIN55212-2 (WIN; 8 mg/kg) acute treatments. Inactivated (p-T286) MEK1, activated (p-S217/221) MEK1/2, activated (p-T202/Y204) ERK1/2, p-S191 FADD, and the corresponding total forms of these proteins were quantified by immunoblotting. RESULTS: Increased (+ 88%) p-T286 MEK1 cortical density, with a concomitant reduction (-43%) of activated ERK was observed in UK-treated mice. Both effects were attenuated by Gαi2 or Gαz antisense ODNs. Contrastingly, WIN induced Gαi2- and Gαz-independent upregulations of p-T286 MEK1 (+ 63%), p-S217/221 MEK1/2 (+ 86%), and activated ERK (+ 111%) in brain. Pro-apoptotic FADD was downregulated (- 34 to 39%) following UK and WIN administration, whereas the neuroprotective p-S191 FADD was increased (+ 74%) in WIN-treated mice only. None of these latter effects required from Gαi2 or Gαz protein integrity. CONCLUSION: The results indicate that α2A-AR (UK), but not CB1-R (WIN), agonists use Gαi2 and Gαz proteins to modulate MEK-ERK, but not FADD, pathway in mouse brain cortex.


Assuntos
Córtex Cerebral/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Subunidade alfa Gi2 de Proteína de Ligação ao GTP/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/fisiologia , Animais , Regulação para Baixo/fisiologia , Masculino , Camundongos , Fosforilação/fisiologia , Regulação para Cima/fisiologia
15.
Schizophr Res ; 215: 493-498, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-28583708

RESUMO

OBJECTIVE: The extracellular matrix protein reelin regulates early brain development and synaptic plasticity in adulthood. Reelin is decreased in the postmortem brain in schizophrenia patients. Reelin's two receptors, ApoER2 and VLDLR, are also substrates for ApoE - a key lipoprotein that regulates phospholipid homeostasis in the brain. The goal of the present study was therefore to examine phospholipids and their constituent fatty acids, and determine whether there is an association between reelin, its receptors and phospholipids in the brain. METHODS: Dorsolateral prefrontal cortex (BA9) grey matter was obtained from the Stanley Foundation Neuropathology Consortium. Samples included tissue from 35 controls, 35 schizophrenia and 34 bipolar disorder patients. Phospholipids were measured using gas liquid chromatography. RESULTS: We quantified 15 individual fatty acid or plasmalogen species for phosphatidylethanolamine and phosphatidylcholine fractions, each comprising >0.5% of the total fatty acid pool. There were no group differences in phospholipids or individual fatty acid species after correcting for multiple comparisons. However, for the entire cohort, both the polyunsaturated subclass of fatty acids, and ApoE, correlated significantly with reelin expression, with a number of individual ω-6 fatty acid species also demonstrating a significant positive correlation. There was a non-significant trend for similar effects with VLDLR expression as for reelin. CONCLUSION: Phospholipids and fatty acids in the dorsolateral cortex do not differ in patients with schizophrenia, bipolar disorder and controls. Reelin expression in this brain region is associated with polyunsaturated fatty acids and ApoE, suggesting further study of potential physiological interactions between these substrates is warranted.


Assuntos
Transtorno Bipolar/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Ácidos Graxos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fosfolipídeos/metabolismo , Córtex Pré-Frontal/metabolismo , Esquizofrenia/metabolismo , Serina Endopeptidases/metabolismo , Adulto , Autopsia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Reelina
16.
Dialogues Clin Neurosci ; 21(3): 271-279, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31749651

RESUMO

Prospective, community-based studies allow evaluation of associations between cognitive functioning and synaptic measures, controlled for age-related pathologies. Findings from >400 community-based participants are reviewed. Levels of two presynaptic proteins, complexin-I (inhibitory terminals), and complexin-II (excitatory terminals) contributed to cognitive variation from normal to dementia. Adding the amount of protein-protein interaction between two others, synaptosome-associated protein-25 and syntaxin, explained 6% of overall variance. The presynaptic protein Munc18-1 long variant was localized to inhibitory terminals, and like complexin-I, was positively associated with cognition. Associations depended on Braak stage, with the level of complexin-I contributing nearly 15% to cognitive variation in stages 0-II, while complexin-II contributed 7% in stages V-VI. Non-denaturing gels identified multiple soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein-protein (SNARE) complexes in frontal and in temporal lobes, making specific contributions to cognitive functions. Multiple mechanisms of presynaptic plasticity contribute to cognitive function during aging.
.


Los estudios prospectivos realizados en la comunidad permiten evaluar las asociaciones entre el funcionamiento cognitivo y las mediciones sinápticas, controladas por patologías relacionadas con la edad. Se revisan los hallazgos de más de 400 participantes de la comunidad. Los niveles de dos proteínas presinápticas, complexina-I (terminales inhibitorios) y complexina-II (terminales excitatorios) contribuyeron a la variación cognitiva entre la situación normal y la demencia. Al agregar la interacción proteína-proteína entre la proteína 25 asociada al sinaptosoma y la sintaxina, se explicó el 6% de la varianza general. La variante larga de la proteína presináptica Munc 18-1 se localizó en terminales inhibitorios y, al igual que la complexina I, se asoció positivamente con la cognición. Las asociaciones dependían de la etapa de Braak, siendo el nivel de complexina-I responsable de casi el 15% de la variación cognitiva para las etapas 0-II, mientras que la complexina-II contribuyó con el 7% en las etapas V-VI. Los geles no desnaturalizantes identificaron múltiples complejos de proteína-proteína del receptor de proteína de unión al factor sensible a N-etilmaleimida soluble (SNARE) en los lóbulos frontales y temporales, los cuales contribuyen de manera específica a las funciones cognitivas. Durante el envejecimiento participan múltiples mecanismos de plasticidad presináptica en la función cognitiva.


Des études prospectives communautaires permettent d'évaluer des associations entre le fonctionnement cognitif et des mesures synaptiques, contrôlées pour des pathologies liées à l'âge. Des résultats issus de plus de 400 participants communautaires sont analysés. Les taux de complexine-I (terminaisons inhibitrices) et de complexine-II (terminaisons excitatrices), deux protéines présynaptiques, contribuent aux variations cognitives de la normalité à la démence. S'y ajoute l'interaction protéine-protéine entre la protéine SNAP25 (protéine 25 associée au synaptosome) et la syntaxine, expliquant 6 % de la variance totale. Le variant long de la protéine pré-synaptique Munc18-1 est localisé sur les terminaisons inhibitrices et associé positivement à la cognition, comme la complexine-I. Les associations dépendent du stade Braak, le taux de complexine-I étant responsable de presque 15 % de la variation cognitive pour les stades 0-II alors que la complexine-II contribue pour 7 % aux stades V-VI. Des gels non dénaturants identifient des complexes multiples protéine-protéine SNARE (Soluble N-ethylmaleimide-sensitive factor Attachment protein REceptor) dans les lobes frontaux et temporaux, contribuant spécifiquement aux fonctions cognitives. Au cours du vieillissement, de nombreux mécanismes de plasticité présynaptique participent à la fonction cognitive.


Assuntos
Cognição/fisiologia , Disfunção Cognitiva/patologia , Proteínas SNARE/fisiologia , Sinapses/patologia , Sinapses/fisiologia , Envelhecimento/patologia , Envelhecimento/psicologia , Animais , Disfunção Cognitiva/psicologia , Humanos , Estudos Prospectivos
17.
Neuroscience ; 420: 97-111, 2019 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-30610939

RESUMO

Abnormalities of SNAP25 (synaptosome-associated protein 25) amount and protein-protein interactions occur in schizophrenia, and may contribute to abnormalities of neurotransmitter release in patients. However, presynaptic terminal function depends on multiple subcellular mechanisms, including energy provided by mitochondria. To explore the SNAP25 interactome in schizophrenia, we immunoprecipitated SNAP25 along with interacting proteins from the ventromedial caudate of 15 cases of schizophrenia and 13 controls. Proteins were identified with mass spectrometry-based proteomics. As well as 15 SNARE- (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) associated proteins, we identified 17 mitochondria-associated and four other proteins. The mitochondrial small GTPase ARF1 (ADP-ribosylation factor 1) was identified in eight schizophrenia SNAP25 immunoprecipitates and none from controls (P = 0.004). Although the ARF1-SNAP25 interaction may be increased, immunoblotting demonstrated 21% lower ARF1-21 (21 kiloDaltons) in schizophrenia samples (P = 0.04). In contrast, the mitochondrial protein UQCRC1 (ubiquinol-cytochrome c reductase core protein 1) did not differ. Lower ARF1-21 levels were associated with the previously reported increased SNAP25-syntaxin interaction in schizophrenia (r = -0.39, P = 0.04). Additional immunoprecipitation studies confirmed the ARF1-21-SNAP25 interaction, independent of UQCRC1. Both ARF1 and SNAP25 were localized to synaptosomes. Confocal microscopy demonstrated co-localization of ARF1 and SNAP25, and further suggested fivefold enrichment of ARF1 in synaptosomes containing an excitatory marker (vesicular glutamate transporter) compared with synaptosomes containing an inhibitory marker (vesicular GABA transporter). The present findings suggest an association between abnormalities of SNARE proteins involved with vesicular neurotransmission and the mitochondrial protein ARF1 that may contribute to the pathophysiology of schizophrenia.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Núcleo Caudado/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatologia , Proteína 25 Associada a Sinaptossoma/metabolismo , Adulto , Idoso , Núcleo Caudado/fisiopatologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
18.
Neuroscience ; 420: 112-128, 2019 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-30579835

RESUMO

Recent studies associated schizophrenia with enhanced functionality of the presynaptic SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex. Altered degradation pathways of the three core SNARE proteins: synaptosomal-associated protein 25 (SNAP25), syntaxin-1 and vesicle-associated membrane protein (VAMP) could contribute to enhanced complex function. To investigate these pathways, we first identified a 15-kDa SNAP25 fragment (f-S25) in human and rat brains, highly enriched in synaptosomal extractions, and mainly attached to cytosolic membranes with low hydrophobicity. The presence of f-S25 is consistent with reports of calpain-mediated SNAP25 cleavage. Co-immunoprecipitation assays showed that f-S25 retains the ability to bind syntaxin-1, which might prevent VAMP and/or Munc18-1 assembly into the complex. Quantitative analyses in postmortem human orbitofrontal cortex (OFC) revealed that schizophrenia (n = 35), but not major depression (n = 15), is associated with lower amounts of f-S25 (-37%, P = 0.027), and greater SNARE protein-protein interactions (35%, P < 0.001), compared with healthy matched controls (n = 28). Enhanced SNARE complex formation was strongly correlated with lower SNAP25 fragmentation rates (R = 0.563, P < 0.001). Statistical mediation analyses supported the hypothesis that reduced f-S25 density could upregulate SNARE fusion events in schizophrenia. Cortical calpain activity in schizophrenia did not differ from controls. f-S25 levels did not correlate with total calpain activity, indicating that if present, schizophrenia-related calpain dysfunction might occur locally at the presynaptic terminals. Overall, the present findings suggest the existence of an endogenous SNARE complex inhibitor related to SNAP25 proteolysis, associated with enhanced SNARE activity in schizophrenia.


Assuntos
Encéfalo/metabolismo , Fragmentos de Peptídeos/metabolismo , Esquizofrenia/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Autopsia , Humanos , Masculino , Pessoa de Meia-Idade , Proteólise , Ratos , Ratos Sprague-Dawley , Proteínas SNARE/metabolismo
19.
Neuropharmacology ; 55(5): 886-99, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18657552

RESUMO

Opioid drugs have been proposed to promote anti-apoptotic signals in brain through inhibition of FADD protein [García-Fuster et al., 2007. Effects of opiate drugs on Fas-associated protein with death domain (FADD) and effector caspases in the rat brain: Regulation by the ERK1/2 MAP kinase pathway. Neuropsychopharmacology 32, 399-411]. FADD phosphorylation by casein kinase Ialpha (CKIalpha) appears to regulate its non-apoptotic activity. This study investigated the effects of opioids on p-FADD in rat brain, as well as various mechanisms that could link opioid receptors with p-FADD, including the modulation of CKIalpha, Galpha(i) proteins and ERK1/2 signaling. In rat, mouse and human brains, various anti-p-FADD antibodies immunodetected the monomeric and oligomeric forms of this protein, irrespective of the antibody origin and specific Ser191 or Ser194 phosphorylation site. Acute mu- and delta-agonists increased, through specific opioid receptor mechanisms, the content of oligomeric and monomeric p-FADD forms in rat cortical homogenates (25-61%) and subcellular compartments, with most relevant effects for sufentanil in membrane (239%) and nucleus (136%). p-FADD induction vanished with repeated (5days) morphine but not SNC-80, and opioid withdrawal induced a new (morphine) or sustained (SNC-80) stimulatory effect (32-33%). The kappa-agonist (-)-U-50488H failed to stimulate p-FADD. Sufentanil reduced CKI protein and kinase activity in the cytosol (30-37%). Morphine, but not SNC-80, augmented CKIalpha in cytosol, membrane and nucleus (36-104%). In contrast to FADD, the ability of SNC-80 to stimulate p-FADD was not sensitive to ERK1/2 blockade. Pertussis toxin did not prevent the opposite effects of SNC-80 on p-FADD and FADD because the toxin by itself markedly altered their basal contents, indicating that FADD could be a novel toxin target. The upregulation of p-FADD induced by mu/delta-agonists could play a relevant role in the anti-apoptotic and/or neuroplastic effects of opioids.


Assuntos
Analgésicos Opioides/farmacologia , Encéfalo/efeitos dos fármacos , Caseína Quinase Ialfa/metabolismo , Proteína de Domínio de Morte Associada a Fas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos , Peso Molecular , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Frações Subcelulares/efeitos dos fármacos
20.
Curr Neuropharmacol ; 15(1): 174-183, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27264948

RESUMO

Chronic prescription of antipsychotics seems to lose its therapeutic benefits in the prevention of recurring psychotic symptoms. In many instances, the occurrence of relapse from initial remission is followed by an increase in dose of the prescribed antipsychotic. The current understanding of why this occurs is still in its infancy, but a controversial idea that has regained attention recently is the notion of iatrogenic dopamine supersensitivity. Studies on cell cultures and animal models have shown that long-term antipsychotic use is linked to both an upregulation of dopamine D<sub>2</sub>-receptors in the striatum and the emergence of enhanced receptor affinity to endogenous dopamine. These findings have been hypothesized to contribute to the phenomenon known as dopamine supersensitivity psychosis (DSP), which has been clinically typified as the foundation of rebound psychosis, drug tolerance, and tardive dyskinesia. The focus of this review is the update of evidence behind the classification of antipsychotic induced DSP and an investigation of its relationship to treatment resistance. Since antipsychotics are the foundation of illness management, a greater understanding of DSP and its prevention may greatly affect patient outcomes.


Assuntos
Antipsicóticos/efeitos adversos , Dopaminérgicos/uso terapêutico , Transtornos Psicóticos/tratamento farmacológico , Transtornos Psicóticos/etiologia , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Dopamina D2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA