Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.696
Filtrar
1.
Sci Rep ; 14(1): 15239, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956130

RESUMEN

Dysbindin-1, a protein encoded by the schizophrenia susceptibility gene DTNBP1, is reduced in the hippocampus of schizophrenia patients. It is expressed in various cellular populations of the brain and implicated in dopaminergic and glutamatergic transmission. To investigate the impact of reduced dysbindin-1 in excitatory cells on hippocampal-associated behaviors and synaptic transmission, we developed a conditional knockout mouse model with deletion of dysbindin-1 gene in CaMKIIα expressing cells. We found that dysbindin-1 reduction in CaMKII expressing cells resulted in impaired spatial and social memories, and attenuation of the effects of glutamate N-methyl-d-asparate receptor (NMDAR) antagonist MK801 on locomotor activity and prepulse inhibition of startle (PPI). Dysbindin-1 deficiency in CaMKII expressing cells also resulted in reduced protein levels of NMDAR subunit GluN1 and GluN2B. These changes were associated with increased expression of immature dendritic spines in basiliar dendrites and abnormalities in excitatory synaptic transmission in the ventral hippocampus. These results highlight the functional relevance of dysbindin-1 in excitatory cells and its implication in schizophrenia-related pathologies.


Asunto(s)
Disbindina , Hipocampo , Ratones Noqueados , Neuronas , Receptores de N-Metil-D-Aspartato , Transmisión Sináptica , Animales , Disbindina/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Hipocampo/metabolismo , Ratones , Neuronas/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/patología , Esquizofrenia/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Masculino , Maleato de Dizocilpina/farmacología , Conducta Animal , Espinas Dendríticas/metabolismo , Proteínas del Tejido Nervioso
2.
Cell Rep Med ; 5(7): 101652, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39019008

RESUMEN

Based on recent genome-wide association studies, it is theorized that altered regulation of autophagy contributes to the pathophysiology of schizophrenia and bipolar disorder. As activity of autophagy-regulatory pathways is controlled by discrete phosphorylation sites on the relevant proteins, phospho-protein profiling is one of the few approaches available for enabling a quantitative assessment of autophagic activity in the brain. Despite this, a comprehensive phospho-protein assessment in the brains of schizophrenia and bipolar disorder subjects is currently lacking. Using this direction, our broad screening identifies an increase in AMP-activated protein kinase (AMPK)-mediated phospho-activation of the pro-autophagy protein beclin-1 solely in the prefrontal cortex of female, but not male, schizophrenia subjects. Using a reverse translational approach, we surprisingly find that this increase in beclin-1 activity facilitates synapse formation and enhances cognition. These findings are interpreted in the context of human studies demonstrating that female schizophrenia subjects have a lower susceptibility to cognitive dysfunction than males.


Asunto(s)
Autofagia , Beclina-1 , Esquizofrenia , Caracteres Sexuales , Esquizofrenia/patología , Esquizofrenia/metabolismo , Esquizofrenia/genética , Humanos , Femenino , Masculino , Beclina-1/metabolismo , Beclina-1/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Fosforilación , Animales , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Sinapsis/metabolismo , Sinapsis/patología , Transducción de Señal , Ratones , Cognición/fisiología , Adulto
3.
Acta Neuropathol Commun ; 12(1): 108, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38943180

RESUMEN

We quantified and determined for the first time the distribution pattern of the neuropeptide NPFF in the human cerebral cortex and subjacent white matter. To do so, we studied n = 9 cases without neurological disorders and n = 22 cases with neurodegenerative diseases, including sporadic amyotrophic lateral sclerosis (ALS, n = 8), Alzheimer's disease (AD, n = 8), Pick's disease (PiD, n = 3), and schizophrenia (n = 3). NPFF-immunopositive cells were located chiefly, but not exclusively, in the superficial white matter and constituted there a subpopulation of white matter interstitial cells (WMIC): Pyramidal-like and multipolar somata predominated in the gyral crowns, whereas bipolar and ovoid somata predominated in the cortex surrounding the sulci. Their sparsely ramified axons were unmyelinated and exhibited NPFF-positive bead-like varicosities. We found significantly fewer NPFF-immunopositive cells in the gray matter of the frontal, cingulate, and superior temporal gyri of both sporadic ALS and late-stage AD patients than in controls, and significantly fewer NPFF-positive cells in the subjacent as well as deep white matter of the frontal gyrus of these patients compared to controls. Notably, the number of NPFF-positive cells was also significantly lower in the hippocampal formation in AD compared to controls. In PiD, NPFF-positive cells were present in significantly lower numbers in the gray and white matter of the cingulate and frontal gyrii in comparison to controls. In schizophrenic patients, lower wNPFF cell counts in the neocortex were significant and global (cingulate, frontal, superior temporal gyrus, medial, and inferior gyri). The precise functions of NPFF-positive cells and their relationship to the superficial corticocortical white matter U-fibers are currently unknown. Here, NPFF immunohistochemistry and expression characterize a previously unrecognized population of cells in the human brain, thereby providing a new entry-point for investigating their physiological and pathophysiological roles.


Asunto(s)
Corteza Cerebral , Enfermedades Neurodegenerativas , Esquizofrenia , Sustancia Blanca , Humanos , Sustancia Blanca/patología , Sustancia Blanca/metabolismo , Masculino , Esquizofrenia/patología , Esquizofrenia/metabolismo , Femenino , Corteza Cerebral/patología , Corteza Cerebral/metabolismo , Anciano , Persona de Mediana Edad , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/metabolismo , Anciano de 80 o más Años , Oligopéptidos , Adulto , Neuronas/patología , Neuronas/metabolismo
4.
J Proteome Res ; 23(7): 2376-2385, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38856018

RESUMEN

Schizophrenia is a severe psychological disorder. The current diagnosis mainly relies on clinical symptoms and lacks laboratory evidence, which makes it very difficult to make an accurate diagnosis especially at an early stage. Plasma protein profiles of schizophrenia patients were obtained and compared with healthy controls using 4D-DIA proteomics technology. Furthermore, 79 DEPs were identified between schizophrenia and healthy controls. GO functional analysis indicated that DEPs were predominantly associated with responses to toxic substances and platelet aggregation, suggesting the presence of metabolic and immune dysregulation in patients with schizophrenia. KEGG pathway enrichment analysis revealed that DEPs were primarily enriched in the chemokine signaling pathway and cytokine receptor interactions. A diagnostic model was ultimately established, comprising three proteins, namely, PFN1, GAPDH and ACTBL2. This model demonstrated an AUC value of 0.972, indicating its effectiveness in accurately identifying schizophrenia. PFN1, GAPDH and ACTBL2 exhibit potential as biomarkers for the early detection of schizophrenia. The findings of our studies provide novel insights into the laboratory-based diagnosis of schizophrenia.


Asunto(s)
Biomarcadores , Profilinas , Proteómica , Esquizofrenia , Esquizofrenia/metabolismo , Esquizofrenia/diagnóstico , Esquizofrenia/sangre , Humanos , Biomarcadores/sangre , Biomarcadores/metabolismo , Proteómica/métodos , Profilinas/metabolismo , Femenino , Masculino , Adulto , Estudios de Casos y Controles , Gliceraldehído-3-Fosfato Deshidrogenasa (Fosforilante)/metabolismo , Persona de Mediana Edad , Proteínas Sanguíneas/análisis , Proteoma/análisis
5.
J Psychopharmacol ; 38(6): 515-525, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38853592

RESUMEN

BACKGROUND: A better understanding of the mechanisms underlying cognitive impairment in schizophrenia is imperative, as it causes poor functional outcomes and a lack of effective treatments. AIMS: This study aimed to investigate the relationships of two proposed main pathophysiology of schizophrenia, altered prefrontal-striatal connectivity and the dopamine system, with cognitive impairment and their interactions. METHODS: Thirty-three patients with schizophrenia and 27 healthy controls (HCs) who are right-handed and matched for age and sex were recruited. We evaluated their cognition, functional connectivity (FC) between the dorsolateral prefrontal cortex (DLPFC)/middle frontal gyrus (MiFG) and striatum, and the availability of striatal dopamine transporter (DAT) using a cognitive battery investigating attention, memory, and executive function, resting-state functional magnetic resonance imaging with group independent component analysis and single-photon emission computed tomography with 99mTc-TRODAT. RESULTS: Patients with schizophrenia exhibited poorer cognitive performance, reduced FC between DLPFC/MiFG and the caudate nucleus (CN) or putamen, decreased DAT availability in the left CN, and decreased right-left DAT asymmetry in the CN compared to HCs. In patients with schizophrenia, altered imaging markers are associated with cognitive impairments, especially the relationship between DLPFC/MiFG-putamen FC and attention and between DAT asymmetry in the CN and executive function. CONCLUSIONS: This study is the first to demonstrate how prefrontal-striatal hypoconnectivity and altered striatal DAT markers are associated with different domains of cognitive impairment in schizophrenia. More research is needed to evaluate their complex relationships and potential therapeutic implications.


Asunto(s)
Disfunción Cognitiva , Cuerpo Estriado , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática , Imagen por Resonancia Magnética , Esquizofrenia , Tomografía Computarizada de Emisión de Fotón Único , Humanos , Masculino , Femenino , Esquizofrenia/fisiopatología , Esquizofrenia/metabolismo , Esquizofrenia/diagnóstico por imagen , Adulto , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/etiología , Disfunción Cognitiva/diagnóstico por imagen , Cuerpo Estriado/metabolismo , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/fisiopatología , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Dopamina/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/diagnóstico por imagen , Corteza Prefrontal/fisiopatología , Corteza Prefontal Dorsolateral/metabolismo , Estudios de Casos y Controles , Persona de Mediana Edad , Función Ejecutiva/fisiología , Pruebas Neuropsicológicas , Adulto Joven
6.
Open Biol ; 14(6): 240063, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38864245

RESUMEN

Frontotemporal lobe abnormalities are linked to neuropsychiatric disorders and cognition, but the role of cellular heterogeneity between temporal lobe (TL) and frontal lobe (FL) in the vulnerability to genetic risk factors remains to be elucidated. We integrated single-nucleus transcriptome analysis in 'fresh' human FL and TL with genetic susceptibility, gene dysregulation in neuropsychiatric disease and psychoactive drug response data. We show how intrinsic differences between TL and FL contribute to the vulnerability of specific cell types to both genetic risk factors and psychoactive drugs. Neuronal populations, specifically PVALB neurons, were most highly vulnerable to genetic risk factors for psychiatric disease. These psychiatric disease-associated genes were mostly upregulated in the TL, and dysregulated in the brain of patients with obsessive-compulsive disorder, bipolar disorder and schizophrenia. Among these genes, GRIN2A and SLC12A5, implicated in schizophrenia and bipolar disorder, were significantly upregulated in TL PVALB neurons and in psychiatric disease patients' brain. PVALB neurons from the TL were twofold more vulnerable to psychoactive drugs than to genetic risk factors, showing the influence and specificity of frontotemporal lobe differences on cell vulnerabilities. These studies provide a cell type resolved map of the impact of brain regional differences on cell type vulnerabilities in neuropsychiatric disorders.


Asunto(s)
Lóbulo Frontal , Trastornos Mentales , Psicotrópicos , Lóbulo Temporal , Humanos , Psicotrópicos/farmacología , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Lóbulo Temporal/metabolismo , Lóbulo Temporal/patología , Trastornos Mentales/genética , Trastornos Mentales/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Predisposición Genética a la Enfermedad , Perfilación de la Expresión Génica , Transcriptoma , Regulación de la Expresión Génica , Esquizofrenia/genética , Esquizofrenia/metabolismo , Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo
7.
Drug Dev Res ; 85(4): e22225, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38879781

RESUMEN

Schizophrenia (SZ) is a serious, destructive neurodevelopmental disorder. Antipsychotic medications are the primary therapy approach for this illness, but it's important to pay attention to the adverse effects as well. Clinical studies for SZ are currently in phase ΙΙΙ for SEP-363856 (SEP-856)-a new antipsychotic that doesn't work on dopamine D2 receptors. However, the underlying action mechanism of SEP-856 remains unknown. This study aimed to evaluate the impact and underlying mechanisms of SEP-856 on SZ-like behavior in a perinatal MK-801 treatment combined with social isolation from the weaning to adulthood model (MK-SI). First, we created an animal model that resembles SZ that combines the perinatal MK-801 with social isolation from weaning to adulthood. Then, different classical behavioral tests were used to evaluate the antipsychotic properties of SEP-856. The levels of proinflammatory cytokines (tumor necrosis factor-α, interleukin-6, and interleukin-1ß), apoptosis-related genes (Bax and Bcl-2), and synaptic plasticity-related genes (brain-derived neurotrophic factor [BDNF] and PSD-95) in the hippocampus were analyzed by quantitative real-time PCR. Hematoxylin and eosin staining were used to observe the morphology of neurons in the hippocampal DG subregions. Western blot was performed to detect the protein expression levels of BDNF, PSD-95, Bax, Bcl-2, PI3K, p-PI3K, AKT, p-AKT, GSK-3ß, p-GSK-3ß in the hippocampus. MK-SI neurodevelopmental disease model studies have shown that compared with sham group, MK-SI group exhibit higher levels of autonomic activity, stereotyped behaviors, withdrawal from social interactions, dysregulated sensorimotor gating, and impaired recognition and spatial memory. These findings imply that the MK-SI model can mimic symptoms similar to those of SZ. Compared with the MK-SI model, high doses of SEP-856 all significantly reduced increased activity, improved social interaction, reduced stereotyping behavior, reversed sensorimotor gating dysregulation, and improved recognition memory and spatial memory impairment in MK-SI mice. In addition, SEP-856 can reduce the release of proinflammatory factors in the MK-SI model, promote the expression of BDNF and PSD-95 in the hippocampus, correct the Bax/Bcl-2 imbalance, turn on the PI3K/AKT/GSK-3ß signaling pathway, and ultimately help the MK-SI mice's behavioral abnormalities. SEP-856 may play an antipsychotic role in MK-SI "dual-hit" model-induced SZ-like behavior mice by promoting synaptic plasticity recovery, decreasing death of hippocampal neurons, lowering the production of pro-inflammatory substances in the hippocampal region, and subsequently initiating the PI3K/AKT/GSK-3ß signaling cascade.


Asunto(s)
Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3 beta , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Esquizofrenia , Transducción de Señal , Animales , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Fármacos Neuroprotectores/farmacología , Antipsicóticos/farmacología , Femenino , Maleato de Dizocilpina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Conducta Animal/efectos de los fármacos , Ratones Endogámicos C57BL , Aislamiento Social
8.
Life Sci Alliance ; 7(9)2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38918041

RESUMEN

Schizophrenia is associated with altered cortical circuitry. Although the schizophrenia risk gene NRG1 is known to affect the wiring of inhibitory interneurons, its role in excitatory neurons and axonal development is unclear. Here, we investigated the role of Nrg1 in the development of the corpus callosum, the major interhemispheric connection formed by cortical excitatory neurons. We found that deletion of Nrg1 impaired callosal axon development in vivo. Experiments in vitro and in vivo demonstrated that Nrg1 is cell-autonomously required for axonal outgrowth and that intracellular signaling of Nrg1 is sufficient to promote axonal development in cortical neurons and specifically in callosal axons. Furthermore, our data suggest that Nrg1 signaling regulates the expression of Growth Associated Protein 43, a key regulator of axonal growth. In conclusion, our study demonstrates that NRG1 is involved in the formation of interhemispheric callosal connections and provides a novel perspective on the relevance of NRG1 in excitatory neurons and in the etiology of schizophrenia.


Asunto(s)
Axones , Cuerpo Calloso , Neurregulina-1 , Transducción de Señal , Animales , Neurregulina-1/metabolismo , Neurregulina-1/genética , Cuerpo Calloso/metabolismo , Axones/metabolismo , Ratones , Esquizofrenia/metabolismo , Esquizofrenia/genética , Esquizofrenia/etiología , Esquizofrenia/patología , Ratones Noqueados , Neuronas/metabolismo , Proteína GAP-43/metabolismo , Proteína GAP-43/genética , Ratones Endogámicos C57BL
9.
J Med Chem ; 67(13): 10831-10847, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38888621

RESUMEN

Selective activation of the M4 muscarinic acetylcholine receptor subtype offers a novel strategy for the treatment of psychosis in multiple neurological disorders. Although the development of traditional muscarinic activators has been stymied due to pan-receptor activation, muscarinic receptor subtype selectivity can be achieved through the utilization of a subtype of a unique allosteric site. A major challenge in capitalizing on this allosteric site to date has been achieving a balance of suitable potency and brain penetration. Herein, we describe the design of a brain penetrant series of M4 selective positive allosteric modulators (PAMs), ultimately culminating in the identification of 21 (PF-06852231, now CVL-231/emraclidine), which is under active clinical development as a novel mechanism and approach for the treatment of schizophrenia.


Asunto(s)
Encéfalo , Diseño de Fármacos , Receptor Muscarínico M4 , Receptor Muscarínico M4/metabolismo , Receptor Muscarínico M4/agonistas , Regulación Alostérica/efectos de los fármacos , Humanos , Animales , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Relación Estructura-Actividad , Ratas , Cricetulus , Células CHO , Agonistas Muscarínicos/farmacología , Agonistas Muscarínicos/síntesis química , Agonistas Muscarínicos/química , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo
10.
Neuroscience ; 551: 205-216, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38843988

RESUMEN

Here, we explored the impact of prolonged environmental enrichment (EE) on behavioral, neurochemical, and epigenetic changes in the serotonin transporter gene in mice subjected to a two-hit schizophrenia model. The methodology involved administering the viral mimetic PolyI:C to neonatal Swiss mice as a first hit during postnatal days (PND) 5-7, or a sterile saline solution as a control. At PND21, mice were randomly assigned either to standard environment (SE) or EE housing conditions. Between PND35-44, the PolyI:C-treated group was submitted to various unpredictable stressors, constituting the second hit. Behavioral assessments were conducted on PND70, immediately after the final EE exposure. Following the completion of behavioral assessments, we evaluated the expression of proteins in the hippocampus that are indicative of microglial activation, such as Iba-1, as well as related to neurogenesis, including doublecortin (Dcx). We also performed methylation analysis on the serotonin transporter gene (Slc6a4) to investigate alterations in serotonin signaling. The findings revealed that EE for 50 days mitigated sensorimotor gating deficits and working memory impairments in two-hit mice and enhanced their locomotor and exploratory behaviors. EE also normalized the overexpression of hippocampal Iba-1 and increased the expression of hippocampal Dcx. Additionally, we observed hippocampal demethylation of the Slc6a4 gene in the EE-exposed two-hit group, indicating epigenetic reprogramming. These results contribute to the growing body of evidence supporting the protective effects of long-term EE in counteracting behavioral disruptions caused by the two-hit schizophrenia model, pointing to enhanced neurogenesis, diminished microglial activation, and epigenetic modifications of serotonergic pathways as underlying mechanisms.


Asunto(s)
Modelos Animales de Enfermedad , Ambiente , Hipocampo , Esquizofrenia , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Animales , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Hipocampo/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/genética , Ratones , Masculino , Proteína Doblecortina , Regiones Promotoras Genéticas , Metilación de ADN , Poli I-C , Neurogénesis/fisiología , Filtrado Sensorial/fisiología
11.
World J Biol Psychiatry ; 25(6): 317-329, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38869228

RESUMEN

OBJECTIVES: Neural stem/progenitor cells derived from olfactory neuroepithelium (hereafter olfactory neural stem/progenitor cells, ONSPCs) are emerging as a potential tool in the exploration of psychiatric disorders. The present study intended to assess whether ONSPCs could help discern individuals with schizophrenia (SZ) from non-schizophrenic (NS) subjects by exploring specific cellular and molecular features. METHODS: ONSPCs were collected from 19 in-patients diagnosed with SZ and 31 NS individuals and propagated in basal medium. Mitochondrial ATP production, expression of ß-catenin and cell proliferation, which are described to be altered in SZ, were examined in freshly isolated or newly thawed ONSPCs after a few culture passages. RESULTS: SZ-ONSPCs exhibited a lower mitochondrial ATP production and insensitivity to agents capable of positively or negatively affecting ß-catenin expression with respect to NS-ONSPCs. As to proliferation, it declined in SZ-ONSPCs as the number of culture passages increased compared to a steady level of growth shown by NS-ONSPCs. CONCLUSIONS: The ease and safety of sample collection as well as the differences observed between NS- and SZ-ONSPCs, may lay the groundwork for a new approach to obtain biological material from a large number of living individuals and gain a better understanding of the mechanisms underlying SZ pathophysiology.


Asunto(s)
Proliferación Celular , Células-Madre Neurales , Mucosa Olfatoria , Esquizofrenia , beta Catenina , Esquizofrenia/metabolismo , Esquizofrenia/patología , Humanos , Adulto , Masculino , Femenino , beta Catenina/metabolismo , Mucosa Olfatoria/citología , Mucosa Olfatoria/metabolismo , Mucosa Olfatoria/patología , Adenosina Trifosfato/metabolismo , Persona de Mediana Edad , Células Cultivadas , Mitocondrias/metabolismo , Células Neuroepiteliales/metabolismo
12.
Psychiatr Genet ; 34(4): 79-85, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38842000

RESUMEN

OBJECTIVE: Exploring the role of microRNAs in the antipsychotic efficacy of electroconvulsive therapy (ECT) will contribute to understanding the underlying mechanism through which ECT exerts its therapeutic effects. The primary objective of this study was to identify microRNA alterations before and after ECT in patients with schizophrenia. METHODS: We compared microarray-based microRNA profiles in peripheral blood from eight patients with schizophrenia before and after ECT and eight healthy controls. Then, we aimed to validate selected differentially expressed microRNAs in 30 patients with schizophrenia following a course of ECT, alongside 30 healthy controls by using quantitative reverse-transcription PCR. RESULTS: Microarray-based expression profiling revealed alterations in 681 microRNAs when comparing pre- and post-ECT samples. Subsequent quantitative reverse-transcription PCR analysis of the selected microRNAs (miR-20a-5p and miR-598) did not reveal any statistical differences between pre- and post-ECT samples nor between pre-ECT samples and those of healthy controls. CONCLUSION: As neuroepigenetic studies on ECT are still in their infancy, the results reported in this study are best interpreted as exploratory outcomes. Additional studies are required to explore the potential epigenetic mechanisms underlying the therapeutic efficacy of ECT.


Asunto(s)
Terapia Electroconvulsiva , MicroARNs , Esquizofrenia , Humanos , Esquizofrenia/genética , Esquizofrenia/terapia , Esquizofrenia/metabolismo , MicroARNs/genética , MicroARNs/sangre , Femenino , Masculino , Adulto , Persona de Mediana Edad , Perfilación de la Expresión Génica/métodos , Estudios de Casos y Controles
14.
Int J Mol Sci ; 25(11)2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38891871

RESUMEN

Until the late 1800s, drug development was a chance finding based on observations and repeated trials and errors. Today, drug development must go through many iterations and tests to ensure it is safe, potent, and effective. This process is a long and costly endeavor, with many pitfalls and hurdles. The aim of the present review article is to explore what is needed for a molecule to move from the researcher bench to the patients' bedside, presented from an industry perspective through the development program of cariprazine. Cariprazine is a relatively novel antipsychotic medication, approved for the treatment of schizophrenia, bipolar mania, bipolar depression, and major depression as an add-on. It is a D3-preferring D3-D2 partial agonist with the highest binding to the D3 receptors compared to all other antipsychotics. Based on the example of cariprazine, there are several key factors that are needed for a molecule to move from the researcher bench to the patients' bedside, such as targeting an unmet medical need, having a novel mechanism of action, and a smart implementation of development plans.


Asunto(s)
Antipsicóticos , Piperazinas , Receptores de Dopamina D3 , Humanos , Antipsicóticos/uso terapéutico , Antipsicóticos/farmacología , Piperazinas/uso terapéutico , Piperazinas/farmacología , Receptores de Dopamina D3/metabolismo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Animales , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/metabolismo , Desarrollo de Medicamentos
15.
Int J Mol Sci ; 25(11)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38892092

RESUMEN

For the past 70 years, the dopamine hypothesis has been the key working model in schizophrenia. This has contributed to the development of numerous inhibitors of dopaminergic signaling and antipsychotic drugs, which led to rapid symptom resolution but only marginal outcome improvement. Over the past decades, there has been limited research on the quantifiable pathological changes in schizophrenia, including premature cellular/neuronal senescence, brain volume loss, the attenuation of gamma oscillations in electroencephalograms, and the oxidation of lipids in the plasma and mitochondrial membranes. We surmise that the aberrant activation of the aryl hydrocarbon receptor by toxins derived from gut microbes or the environment drives premature cellular and neuronal senescence, a hallmark of schizophrenia. Early brain aging promotes secondary changes, including the impairment and loss of mitochondria, gray matter depletion, decreased gamma oscillations, and a compensatory metabolic shift to lactate and lactylation. The aim of this narrative review is twofold: (1) to summarize what is known about premature cellular/neuronal senescence in schizophrenia or schizophrenia-like disorders, and (2) to discuss novel strategies for improving long-term outcomes in severe mental illness with natural senotherapeutics, membrane lipid replacement, mitochondrial transplantation, microbial phenazines, novel antioxidant phenothiazines, inhibitors of glycogen synthase kinase-3 beta, and aryl hydrocarbon receptor antagonists.


Asunto(s)
Antipsicóticos , Esquizofrenia , Humanos , Antipsicóticos/uso terapéutico , Antipsicóticos/farmacología , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Trastornos Psicóticos/tratamiento farmacológico , Trastornos Psicóticos/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Senescencia Celular/efectos de los fármacos
16.
Biomed Pharmacother ; 175: 116747, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38744217

RESUMEN

Schizophrenia, influenced by genetic and environmental factors, may involve epigenetic alterations, notably histone modifications, in its pathogenesis. This review summarizes various histone modifications including acetylation, methylation, phosphorylation, ubiquitination, serotonylation, lactylation, palmitoylation, and dopaminylation, and their implications in schizophrenia. Current research predominantly focuses on histone acetylation and methylation, though other modifications also play significant roles. These modifications are crucial in regulating transcription through chromatin remodeling, which is vital for understanding schizophrenia's development. For instance, histone acetylation enhances transcriptional efficiency by loosening chromatin, while increased histone methyltransferase activity on H3K9 and altered histone phosphorylation, which reduces DNA affinity and destabilizes chromatin structure, are significant markers of schizophrenia.


Asunto(s)
Histonas , Esquizofrenia , Esquizofrenia/metabolismo , Esquizofrenia/genética , Humanos , Histonas/metabolismo , Animales , Epigénesis Genética , Procesamiento Proteico-Postraduccional , Acetilación , Metilación , Fosforilación , Ensamble y Desensamble de Cromatina
17.
Behav Brain Res ; 470: 115053, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-38768688

RESUMEN

BACKGROUND: Adverse experiences due to early life stress (ELS) or parental psychopathology such as schizophrenia (SZ) have a significant implication on individual susceptibility to psychiatric disorders in the future. However, it is not fully understood how ELS affects social-associated behaviors as well as the developing prefrontal cortex (PFC). OBJECTIVE: The aim of this study was to investigate the impact of ELS and ketamine induced schizophrenia like symptoms (KSZ) on anhedonia, social behavior and anxiety-like behavior. METHODS: Male and female Sprague-Dawley rat pups were allocated randomly into eight experimental groups, namely control, gestational stress (GS), GS+KSZ, maternal separation (MS), MS+KSZ pups, KSZ parents, KSZ parents and Pups and KSZ pups only. ELS was induced by subjecting the pups to GS and MS, while schizophrenia like symptoms was induced through subcutaneous administration of ketamine. Behavioral assessment included sucrose preference test (SPT) and elevated plus maze (EPM), followed by dopamine testing and analysis of astrocyte density. Statistical analysis involved ANOVA and post hoc Tukey tests, revealing significant group differences and yielding insights into behavioral and neurodevelopmental impacts. RESULTS: GS, MS, and KSZ (dams) significantly reduced hedonic response and increased anxiety-like responses (p < 0.05). Notably, the presence of normal parental mental health demonstrated a reversal of the observed decline in Glial Fibrillary Acidic Protein-positive astrocytes (GFAP+ astrocytes) (p < 0.05) and a reduction in anxiety levels, implying its potential protective influence on depressive-like symptoms and PFC astrocyte functionality. CONCLUSION: The present study provides empirical evidence supporting the hypothesis that exposure to ELS and KSZ on dams have a significant impact on the on development of anxiety and depressive like symptoms in Sprague Dawley rats, while positive parenting has a reversal effect.


Asunto(s)
Ansiedad , Depresión , Ketamina , Privación Materna , Corteza Prefrontal , Ratas Sprague-Dawley , Esquizofrenia , Estrés Psicológico , Animales , Femenino , Esquizofrenia/metabolismo , Esquizofrenia/inducido químicamente , Ketamina/farmacología , Masculino , Ratas , Corteza Prefrontal/metabolismo , Embarazo , Modelos Animales de Enfermedad , Efectos Tardíos de la Exposición Prenatal , Conducta Animal/fisiología , Anhedonia/fisiología , Conducta Social , Psicología del Esquizofrénico , Dopamina/metabolismo , Astrocitos/metabolismo
18.
Sci Transl Med ; 16(749): eadh9974, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38781321

RESUMEN

Many psychiatric disorders exhibit sex differences, but the underlying mechanisms remain poorly understood. We analyzed transcriptomics data from 2160 postmortem adult prefrontal cortex brain samples from the PsychENCODE consortium in a sex-stratified study design. We compared transcriptomics data of postmortem brain samples from patients with schizophrenia (SCZ), bipolar disorder (BD), and autism spectrum disorder (ASD) with transcriptomics data of postmortem control brains from individuals without a known history of psychiatric disease. We found that brain samples from females with SCZ, BD, and ASD showed a higher burden of transcriptomic dysfunction than did brain samples from males with these disorders. This observation was supported by the larger number of differentially expressed genes (DEGs) and a greater magnitude of gene expression changes observed in female versus male brain specimens. In addition, female patient brain samples showed greater overall connectivity dysfunction, defined by a higher proportion of gene coexpression modules with connectivity changes and higher connectivity burden, indicating a greater degree of gene coexpression variability. We identified several gene coexpression modules enriched in sex-biased DEGs and identified genes from a genome-wide association study that were involved in immune and synaptic functions across different brain cell types. We found a number of genes as hubs within these modules, including those encoding SCN2A, FGF14, and C3. Our results suggest that in the context of psychiatric diseases, males and females exhibit different degrees of transcriptomic dysfunction and implicate immune and synaptic-related pathways in these sex differences.


Asunto(s)
Autopsia , Encéfalo , Trastornos Mentales , Caracteres Sexuales , Transcriptoma , Humanos , Femenino , Masculino , Transcriptoma/genética , Encéfalo/metabolismo , Encéfalo/patología , Trastornos Mentales/genética , Trastornos Mentales/patología , Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo , Trastorno Bipolar/patología , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patología , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Adulto , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Redes Reguladoras de Genes , Persona de Mediana Edad
19.
Schizophr Res ; 269: 58-63, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38733800

RESUMEN

N-acetylasparate and lactate are two prominent brain metabolites closely related to mitochondrial functioning. Prior research revealing lower levels of NAA and higher levels of lactate in the cerebral cortex of patients with schizophrenia suggest possible abnormalities in the energy supply pathway necessary for brain function. Given that stress and adversity are a strong risk factor for a variety of mental health problems, including psychotic disorders, we investigated the hypothesis that stress contributes to abnormal neuroenergetics in patients with schizophrenia. To test this hypothesis, we used the Stress and Adversity Inventory (STRAIN) to comprehensively assess the lifetime stressor exposure profiles of 35 patients with schizophrenia spectrum disorders and 33 healthy controls who were also assessed with proton magnetic resonance spectroscopy at the anterior cingulate cortex using 3 Tesla scanner. Consistent with the hypothesis, greater lifetime stressor exposure was significantly associated with lower levels of N-acetylasparate (ß = -0.36, p = .005) and higher levels of lactate (ß = 0.43, p = .001). Moreover, these results were driven by patients, as these associations were significant for the patient but not control group. Though preliminary, these findings suggest a possible role for stress processes in the pathophysiology of abnormal neuroenergetics in schizophrenia.


Asunto(s)
Ácido Aspártico , Ácido Láctico , Esquizofrenia , Estrés Psicológico , Humanos , Masculino , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Esquizofrenia/diagnóstico por imagen , Femenino , Adulto , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Ácido Láctico/metabolismo , Ácido Láctico/sangre , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Espectroscopía de Protones por Resonancia Magnética , Persona de Mediana Edad , Adulto Joven , Trastornos Psicóticos/metabolismo , Trastornos Psicóticos/fisiopatología , Trastornos Psicóticos/diagnóstico por imagen , Giro del Cíngulo/metabolismo , Giro del Cíngulo/diagnóstico por imagen , Giro del Cíngulo/fisiopatología , Espectroscopía de Resonancia Magnética
20.
Nat Commun ; 15(1): 3980, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730231

RESUMEN

Schizophrenia is a complex neuropsychiatric disorder with sexually dimorphic features, including differential symptomatology, drug responsiveness, and male incidence rate. Prior large-scale transcriptome analyses for sex differences in schizophrenia have focused on the prefrontal cortex. Analyzing BrainSeq Consortium data (caudate nucleus: n = 399, dorsolateral prefrontal cortex: n = 377, and hippocampus: n = 394), we identified 831 unique genes that exhibit sex differences across brain regions, enriched for immune-related pathways. We observed X-chromosome dosage reduction in the hippocampus of male individuals with schizophrenia. Our sex interaction model revealed 148 junctions dysregulated in a sex-specific manner in schizophrenia. Sex-specific schizophrenia analysis identified dozens of differentially expressed genes, notably enriched in immune-related pathways. Finally, our sex-interacting expression quantitative trait loci analysis revealed 704 unique genes, nine associated with schizophrenia risk. These findings emphasize the importance of sex-informed analysis of sexually dimorphic traits, inform personalized therapeutic strategies in schizophrenia, and highlight the need for increased female samples for schizophrenia analyses.


Asunto(s)
Núcleo Caudado , Corteza Prefontal Dorsolateral , Hipocampo , Sitios de Carácter Cuantitativo , Esquizofrenia , Caracteres Sexuales , Humanos , Esquizofrenia/genética , Esquizofrenia/metabolismo , Femenino , Masculino , Hipocampo/metabolismo , Núcleo Caudado/metabolismo , Corteza Prefontal Dorsolateral/metabolismo , Adulto , Transcriptoma , Perfilación de la Expresión Génica , Factores Sexuales , Cromosomas Humanos X/genética , Corteza Prefrontal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...