Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
EMBO J ; 43(8): 1420-1444, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38528182

RESUMEN

Current approaches to the treatment of schizophrenia have mainly focused on the protein-coding part of the genome; in this context, the roles of microRNAs have received less attention. In the present study, we analyze the microRNAome in the blood and postmortem brains of schizophrenia patients, showing that the expression of miR-99b-5p is downregulated in both the prefrontal cortex and blood of patients. Lowering the amount of miR-99b-5p in mice leads to both schizophrenia-like phenotypes and inflammatory processes that are linked to synaptic pruning in microglia. The microglial miR-99b-5p-supressed inflammatory response requires Z-DNA binding protein 1 (Zbp1), which we identify as a novel miR-99b-5p target. Antisense oligonucleotides against Zbp1 ameliorate the pathological effects of miR-99b-5p inhibition. Our findings indicate that a novel miR-99b-5p-Zbp1 pathway in microglia might contribute to the pathogenesis of schizophrenia.


Asunto(s)
MicroARNs , Esquizofrenia , Animales , Humanos , Ratones , Microglía/metabolismo , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Esquizofrenia/genética
2.
Acta Neuropathol ; 147(1): 45, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38407651

RESUMEN

Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease caused by repetitive head impacts (RHI) and pathologically defined as neuronal phosphorylated tau aggregates around small blood vessels and concentrated at sulcal depths. Cross-sectional studies suggest that tau inclusions follow a stereotyped pattern that begins in the neocortex in low stage disease, followed by involvement of the medial temporal lobe and subcortical regions with significant neocortical burden in high stage CTE. Here, we define a subset of brain donors with high stage CTE and with a low overall cortical burden of tau inclusions (mean semiquantitative value ≤1) and classify them as cortical-sparing CTE (CSCTE). Of 620 brain donors with pathologically diagnosed CTE, 66 (11%) met criteria for CSCTE. Compared to typical high stage CTE, those with CSCTE had a similar age at death and years of contact sports participation and were less likely to carry apolipoprotein ε4 (p < 0.05). CSCTE had less overall tau pathology severity, but a proportional increase of disease burden in medial temporal lobe and brainstem regions compared to the neocortex (p's < 0.001). CSCTE also had lower prevalence of comorbid neurodegenerative disease. Clinically, CSCTE participants were less likely to have dementia (p =  0.023) and had less severe cognitive difficulties (as reported by informants using the Functional Activities Questionnaire (FAQ); p < 0.001, meta-cognitional index T score; p = 0.002 and Cognitive Difficulties Scale (CDS); p < 0.001,) but had an earlier onset age of behavioral (p = 0.006) and Parkinsonian motor (p = 0.013) symptoms when compared to typical high stage CTE. Other comorbid tauopathies likely contributed in part to these differences: when cases with concurrent Alzheimer dementia or frontal temporal lobar degeneration with tau pathology were excluded, differences were largely retained, but only remained significant for FAQ (p = 0.042), meta-cognition index T score (p = 0.014) and age of Parkinsonian motor symptom onset (p = 0.046). Overall, CSCTE appears to be a distinct subtype of high stage CTE with relatively greater involvement of subcortical and brainstem regions and less severe cognitive symptoms.


Asunto(s)
Enfermedad de Alzheimer , Encefalopatía Traumática Crónica , Enfermedades Neurodegenerativas , Humanos , Estudios Transversales , Encéfalo
3.
Proc Natl Acad Sci U S A ; 120(9): e2204933120, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36812208

RESUMEN

N6-methyladenosine (m6A) regulates mRNA metabolism. While it has been implicated in the development of the mammalian brain and in cognition, the role of m6A in synaptic plasticity, especially during cognitive decline, is not fully understood. In this study, we employed methylated RNA immunoprecipitation sequencing to obtain the m6A epitranscriptome of the hippocampal subregions CA1, CA3, and the dentate gyrus and the anterior cingulate cortex (ACC) in young and aged mice. We observed a decrease in m6A levels in aged animals. Comparative analysis of cingulate cortex (CC) brain tissue from cognitively intact human subjects and Alzheimer's disease (AD) patients showed decreased m6A RNA methylation in AD patients. m6A changes common to brains of aged mice and AD patients were found in transcripts linked to synaptic function including calcium/calmodulin-dependent protein kinase 2 (CAMKII) and AMPA-selective glutamate receptor 1 (Glua1). We used proximity ligation assays to show that reduced m6A levels result in decreased synaptic protein synthesis as exemplified by CAMKII and GLUA1. Moreover, reduced m6A levels impaired synaptic function. Our results suggest that m6A RNA methylation controls synaptic protein synthesis and may play a role in cognitive decline associated with aging and AD.


Asunto(s)
Enfermedad de Alzheimer , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Humanos , Ratones , Animales , Anciano , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Hipocampo/metabolismo , Enfermedad de Alzheimer/metabolismo , Envejecimiento/metabolismo , ARN/metabolismo , Mamíferos/genética
4.
Eur Arch Psychiatry Clin Neurosci ; 273(6): 1279-1293, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36302978

RESUMEN

Over the last few years, extracellular vesicles (EVs) have received increasing attention as potential non-invasive diagnostic and therapeutic biomarkers for various diseases. The interest in EVs is related to their structure and content, as well as to their changing cargo in response to different stimuli. One of the potential areas of use of EVs as biomarkers is the central nervous system (CNS), in particular the brain, because EVs can cross the blood-brain barrier, exist also in peripheral tissues and have a diverse cargo. Thus, they may represent "liquid biopsies" of the CNS that can reflect brain pathophysiology without the need for invasive surgical procedures. Overall, few studies to date have examined EVs in neuropsychiatric disorders, and the present evidence appears to lack reproducibility. This situation might be due to a variety of technical obstacles related to working with EVs, such as the use of different isolation strategies, which results in non-uniform vesicular and molecular outputs. Multi-omics approaches and improvements in the standardization of isolation procedures will allow highly pure EV fractions to be obtained in which the molecular cargo, particularly microRNAs and proteins, can be identified and accurately quantified. Eventually, these advances will enable researchers to decipher disease-relevant molecular signatures of the brain-derived EVs involved in synaptic plasticity, neuronal development, neuro-immune communication, and other related pathways. This narrative review summarizes the findings of studies on EVs in major psychiatric disorders, particularly in the field of biomarkers, and discusses the respective therapeutic potential of EVs.


Asunto(s)
Vesículas Extracelulares , Trastornos Mentales , Humanos , Reproducibilidad de los Resultados , Vesículas Extracelulares/metabolismo , Encéfalo , Biomarcadores/metabolismo , Trastornos Mentales/diagnóstico , Trastornos Mentales/terapia , Trastornos Mentales/metabolismo
5.
J Neuropathol Exp Neurol ; 81(12): 1008-1017, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36179256

RESUMEN

Meningioma is the most common intracranial neoplasm, yet there is no effective therapy for recurrent/refractory meningiomas after surgery and radiation. Prostate-specific membrane antigen (PSMA) is an enzyme upregulated on endothelial cells of multiple neoplasms and is being investigated as a theranostic target. Until now, PSMA has not been studied in meningiomas. We aimed to verify PSMA endothelial expression in meningiomas, detect tumor grade variability, and investigate the relationship of PSMA signal with tumor recurrence. We analyzed 96 archival meningiomas including 58 de novo and 38 recurrent specimens. All specimens were stained routinely and immunostained for CD31 and PSMA. Slides were scanned and analyzed producing raw data for images of PSMA, CD31, PSMA/CD31, and PSMA/vasculature. PSMA expression was seen within 98.9% of meningioma samples. In the total cohort, higher-grade tumors had increased expression of raw PSMA and PSMA/CD31, and PSMA/vasculature ratios compared to grade 1 tumors. PSMA expression and PSMA/vasculature ratios (p = 0.0015) were higher in recurrent versus de novo tumors among paired samples. ROC curves demonstrated PSMA/CD31, PSMA/vasculature, and raw CD31 as indicators of tumor recurrence. Thus, PSMA is expressed within endothelial cells of meningiomas, is increased with tumor grade and recurrence, and persists with prior irradiation.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Masculino , Meningioma/cirugía , Recurrencia Local de Neoplasia , Medicina de Precisión , Células Endoteliales , Próstata , Neoplasias Meníngeas/cirugía
7.
Psychiatr Danub ; 33(Suppl 4): 451-455, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34718264

RESUMEN

Bipolar disorder (BD) is a common, recurring psychiatric illness with unknown pathogenesis. Much like other psychiatric diseases, BD suffers from the chronic lack of reliable biomarkers and innovative pharmacological interventions. Better characterization of clinical profiles, experimental medicine, genomic data mining, and the utilization of experimental models, including stem cell and genetically modified mice, are suggested ways forward. Environment, including early childhood experiences, has been documented to modulate the risk for the development of psychiatric disorders via epigenetic mechanisms. Key epigenetic regulators, microRNAs (miRNAs, miRs), govern normal neuronal functioning and show altered expression in diverse brain pathologies. We observed significant alterations of exosomal miR-29c levels in prefrontal cortex (Brodmann area 9, BA9) of BD patients. We also demonstrated that exosomes extracted from the anterior cingulate cortex (BA24), a crucial area for modulating emotional expression and affect, have increased levels of miR-149 in BD patients compared to controls. Because miR-149 has been shown to inhibit glial proliferation, we hypothesized that increased miR-149 expression in BA24-derived exosomes may be consistent with the previously reported reduced glial cell numbers in BA24 of patients diagnosed with familial BD. qPCR analysis of laser-microdissected neuronal and glial cells from BA24 cortical samples of BD patients verified that the glial, but not neuronal, population exhibits significantly increased miR-149 expression. These findings support neuron-glia interaction as a possible target mechanism in BD, implicated by others in neuroimaging, postmortem, and in vivo studies of the pathological changes mediated by glial cells.


Asunto(s)
Trastorno Bipolar , MicroARNs , Animales , Biomarcadores , Trastorno Bipolar/genética , Preescolar , Humanos , Ratones , MicroARNs/genética , Neuronas , Corteza Prefrontal
8.
Noncoding RNA ; 7(1)2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33535543

RESUMEN

The neurological damage of Alzheimer's disease (AD) is thought to be irreversible upon onset of dementia-like symptoms, as it takes years to decades for occult pathologic changes to become symptomatic. It is thus necessary to identify individuals at risk for the development of the disease before symptoms manifest in order to provide early intervention. Surrogate markers are critical for early disease detection, stratification of patients in clinical trials, prediction of disease progression, evaluation of response to treatment, and also insight into pathomechanisms. Here, we review the evidence for a number of microRNAs that may serve as biomarkers with possible mechanistic insights into the AD pathophysiologic processes, years before the clinical manifestation of the disease.

9.
Cereb Cortex Commun ; 1(1): tgaa031, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32974611

RESUMEN

Cerebral amyloid angiopathy (CAA) in Alzheimer's disease (AD)-deposition of beta amyloid (Aß) within the walls of cerebral blood vessels-typically accompanies Aß buildup in brain parenchyma and causes abnormalities in vessel structure and function. We recently demonstrated that the immunoreactivity of activin receptor-like kinase 1 (ALK1), the type I receptor for circulating BMP9/BMP10 (bone morphogenetic protein) signaling proteins, is reduced in advanced, but not early stages of AD in CA3 pyramidal neurons. Here we characterize vascular expression of ALK1 in the context of progressive AD pathology accompanied by amyloid angiopathy in postmortem hippocampi using immunohistochemical methods. Hippocampal arteriolar wall ALK1 signal intensity was 35% lower in AD patients (Braak and Braak Stages IV and V [BBIV-V]; clinical dementia rating [CDR1-2]) as compared with subjects with early AD pathologic changes but either cognitively intact or with minimal cognitive impairment (BBIII; CDR0-0.5). The intensity of Aß signal in arteriolar walls was similar in all analyzed cases. These data suggest that, as demonstrated previously for specific neuronal populations, ALK1 expression in blood vessels is also vulnerable to the AD pathophysiologic process, perhaps related to CAA. However, cortical arterioles may remain responsive to the ALK1 ligands, such as BMP9 and BMP10 in early and moderate AD.

10.
Commun Biol ; 2: 360, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31602409

RESUMEN

Alzheimer's disease (AD) is a progressive, neurodegenerative dementia with no cure. Prominent hypotheses suggest accumulation of beta-amyloid (Aß) contributes to neurodegeneration and memory loss, however identifying brain regions with early susceptibility to Aß remains elusive. Using SWITCH to immunolabel intact brain, we created a spatiotemporal map of Aß deposition in the 5XFAD mouse. We report that subcortical memory structures show primary susceptibility to Aß and that aggregates develop in increasingly complex networks with age. The densest early Aß occurs in the mammillary body, septum, and subiculum- core regions of the Papez memory circuit. Previously, early mammillary body dysfunction in AD had not been established. We also show that Aß in the mammillary body correlates with neuronal hyper-excitability and that modulation using a pharmacogenetic approach reduces Aß deposition. Our data demonstrate large-tissue volume processing techniques can enhance biological discovery and suggest that subcortical susceptibility may underlie early brain alterations in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Enfermedad de Alzheimer/patología , Amiloidosis/metabolismo , Amiloidosis/patología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Ratones Transgénicos
11.
Transl Psychiatry ; 9(1): 250, 2019 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-31591382

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder causing huge emotional and economic burden to our societies. An effective therapy has not been implicated yet, which is in part also due to the fact that pathological changes occur years before clinical symptoms manifest. Thus, there is a great need for the development of a translatable biomarker. Recent evidence highlights microRNAs as candidate biomarkers. In this study, we use next-generation sequencing to study the small noncoding RNAome (sncRNAome) in exosomes derived from human cerebrospinal fluid (CSF). We show that the sncRNAome from CSF-derived exosomes is dominated not only by microRNAs (miRNAs) but also by PIWI-interacting RNAs (piRNAs). We define a combined signature consisting of three miRNAs and three piRNAs that are suitable to detect AD with an AUC of 0.83 in a replication cohort and furthermore predict the conversion of mild-cognitive impaired (MCI) patients to AD dementia with an AUC of 0.86 for the piRNA signature. When combining the smallRNA signature with pTau and Aß 42/40 ratio the AUC reaches 0.98. Our study reports a novel exosomal small noncoding RNA signature to detect AD pathology and provides the first evidence that in addition to miRNAs, piRNAs should also be considered as a candidate biomarker for AD.


Asunto(s)
Enfermedad de Alzheimer/líquido cefalorraquídeo , Péptidos beta-Amiloides/líquido cefalorraquídeo , Disfunción Cognitiva/líquido cefalorraquídeo , MicroARNs/líquido cefalorraquídeo , Fragmentos de Péptidos/líquido cefalorraquídeo , ARN Interferente Pequeño/líquido cefalorraquídeo , Adulto , Anciano , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Área Bajo la Curva , Biomarcadores/líquido cefalorraquídeo , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/genética , Estudios de Cohortes , Femenino , Alemania , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , ARN Interferente Pequeño/genética
12.
Cureus ; 11(5): e4726, 2019 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-31355086

RESUMEN

This case of a radiation-naive patient with pilocytic astrocytoma highlights how deletions of CDKN2A (cyclin-dependent kinase Inhibitor 2A) and PTEN (phosphatase and tensin homolog) portended a poor clinical outcome. Pilocytic astrocytomas are grade 1 tumors usually occurring in children and young adults with KIAA1549-BRAF fusion defining the majority of pilocytic astrocytomas. The presence of CDKN2A and PTEN loss may be associated with aggressive biology in pilocytic astrocytoma and further studies should include comprehensive genomics in a larger series of adult pilocytic astrocytoma to evaluate this previously unreported finding. Providers need to be aware of this possibility given the potential for poor outcomes.

13.
J Alzheimers Dis ; 68(1): 357-365, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30775993

RESUMEN

Genome-wide association studies identified a single nucleotide polymorphism (SNP) in the MSRB3 gene encoding Methionine Sulfoxide Reductase-B3 (MsrB3) to be associated with the risk for low hippocampal volume and late onset Alzheimer's disease (AD). Subsequently, we identified AD-associated abnormal patterns of neuronal and vascular MsrB3 expression in postmortem hippocampi. The present study investigated the relationship between the MSRB3 SNP rs61921502, G (minor/risk allele) and MRI measures of brain injury including total brain volume, hippocampal volume, and white matter hyperintensities using linear regression models; the presence of brain infarcts using logistic regression models; and the incidence of stroke, dementia, and AD using Cox proportional hazards models in 2,038 Framingham Heart Study Offspring participants with MRI administered close to examination cycle 7 (1998-2001). Participants with neurological conditions that impede evaluation of vascular pathology by MRI, i.e., brain tumors, multiple sclerosis, and major head trauma, were excluded from the study. When adjusted for age and age squared at MRI exam, sex, and presence of Apolipoproteinɛ4 allele (APOE4), individuals with MSRB3 rs61921502 minor allele had increased odds for brain infarcts on MRI compared to those with no minor allele. However, in stratified analyses, MSRB3 rs61921502 minor allele was significantly associated with increased odds for MRI brain infarcts only in the absence of APOE4.


Asunto(s)
Alelos , Infarto Encefálico/genética , Demencia/genética , Predisposición Genética a la Enfermedad , Hipocampo/diagnóstico por imagen , Metionina Sulfóxido Reductasas/genética , Polimorfismo de Nucleótido Simple , Anciano , Infarto Encefálico/diagnóstico por imagen , Infarto Encefálico/epidemiología , Demencia/diagnóstico por imagen , Demencia/epidemiología , Femenino , Humanos , Incidencia , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad
14.
J Alzheimers Dis ; 63(4): 1433-1443, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29843236

RESUMEN

The pathophysiology of Alzheimer's disease (AD) includes signaling defects mediated by the transforming growth factor ß-bone morphogenetic protein-growth and differentiation factor (TGFß-BMP-GDF) family of proteins. In animal models of AD, administration of BMP9/GDF2 improves memory and reduces amyloidosis. The best characterized type I receptor of BMP9 is ALK1. We characterized ALK1 expression in the hippocampus using immunohistochemistry. In the rat, ALK1 immunoreactivity was found in CA pyramidal neurons, most frequently and robustly in the CA2 and CA3 fields. In addition, there were sporadic ALK1-immunoreactive cells in the stratum oriens, mainly in CA1. The ALK1 expression pattern in human hippocampus was similar to that of rat. Pyramidal neurons within the CA2, CA3, and CA4 were strongly ALK1-immunoreactive in hippocampi of cognitively intact subjects with no neurofibrillary tangles. ALK1 signal was found in the axons of alveus and fimbria, and in the neuropil across CA fields. Relatively strongest ALK1 neuropil signal was observed in CA1 where pyramidal neurons were occasionally ALK1-immunoractive. As in the rat, horizontally oriented neurons in the stratum oriens of CA1 were both ALK1- and GAD67-immunoreactive. Analysis of ALK1 immunoreactivity across stages of AD pathology revealed that disease progression was characterized by overall reduction of the ALK1 signal in CA3 in advanced, but not early, stages of AD. These data suggest that the CA3 pyramidal neurons may remain responsive to the ALK1 ligands, e.g., BMP9, during initial stages of AD and that ALK1 may constitute a therapeutic target in early and moderate AD.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Receptores de Activinas/metabolismo , Enfermedad de Alzheimer/patología , Región CA3 Hipocampal/metabolismo , Progresión de la Enfermedad , Anciano , Enfermedad de Alzheimer/metabolismo , Animales , Femenino , Glutamato Descarboxilasa/metabolismo , Factor 2 de Diferenciación de Crecimiento , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ratas , Ratas Wistar
15.
J Alzheimers Dis ; 60(1): 43-56, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28777754

RESUMEN

Genome-wide association studies (GWAS) identified susceptibility loci associated with decreased hippocampal volume, and found hippocampal subfield-specific effects at MSRB3 (methionine sulfoxide reductase-B3). The MSRB3 locus was also linked to increased risk for late onset Alzheimer's disease (AD). In this study, we uncovered novel sites of MsrB3 expression in CA pyramidal layer and arteriolar walls by using automated immunohistochemistry on hippocampal sections from 23 individuals accompanied by neuropathology reports and clinical dementia rating scores. Controls, cognitively intact subjects with no hippocampal neurofibrillary tangles, exhibited MsrB3 signal as distinct but rare puncta in CA1 pyramidal neuronal somata. In CA3, however, MsrB3-immunoreactivity was strongest in the neuropil of the pyramidal layer. These patterns were replicated in rodent hippocampi where ultrastructural and immunohistofluorescence analysis revealed MsrB3 signal associated with synaptic vesicles and colocalized with mossy fiber terminals. In AD subjects, the number of CA1 pyramidal neurons with frequent, rather than rare, MsrB3-immunoreactive somatic puncta increased in comparison to controls. This change in CA1 phenotype correlated with the occurrence of AD pathological hallmarks. Moreover, the intensity of MsrB3 signal in the neuropil of CA3 pyramidal layer correlated with the signal pattern in neurons of CA1 pyramidal layer that was characteristic of cognitively intact individuals. Finally, MsrB3 signal in the arteriolar walls in the hippocampal white matter decreased in AD patients. This characterization of GWAS-implicated MSRB3 protein expression in human hippocampus suggests that patterns of neuronal and vascular MsrB3 protein expression reflect or underlie pathology associated with AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Hipocampo/metabolismo , Hipocampo/patología , Metionina Sulfóxido Reductasas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Animales , Plexo Coroideo/metabolismo , Plexo Coroideo/patología , Plexo Coroideo/ultraestructura , Epéndimo/metabolismo , Epéndimo/patología , Epéndimo/ultraestructura , Femenino , Regulación de la Expresión Génica/fisiología , Estudio de Asociación del Genoma Completo , Hipocampo/ultraestructura , Humanos , Masculino , Metionina Sulfóxido Reductasas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Persona de Mediana Edad , Células Piramidales/metabolismo , Células Piramidales/patología , Células Piramidales/ultraestructura , Ratas , Ratas Wistar , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo
16.
EMBO J ; 36(19): 2815-2828, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28768717

RESUMEN

Age-associated memory decline is due to variable combinations of genetic and environmental risk factors. How these risk factors interact to drive disease onset is currently unknown. Here we begin to elucidate the mechanisms by which post-traumatic stress disorder (PTSD) at a young age contributes to an increased risk to develop dementia at old age. We show that the actin nucleator Formin 2 (Fmn2) is deregulated in PTSD and in Alzheimer's disease (AD) patients. Young mice lacking the Fmn2 gene exhibit PTSD-like phenotypes and corresponding impairments of synaptic plasticity, while the consolidation of new memories is unaffected. However, Fmn2 mutant mice develop accelerated age-associated memory decline that is further increased in the presence of additional risk factors and is mechanistically linked to a loss of transcriptional homeostasis. In conclusion, our data present a new approach to explore the connection between AD risk factors across life span and provide mechanistic insight to the processes by which neuropsychiatric diseases at a young age affect the risk for developing dementia.


Asunto(s)
Demencia/genética , Proteínas de Microfilamentos/genética , Proteínas Nucleares/genética , Adulto , Edad de Inicio , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Estudios de Casos y Controles , Demencia/epidemiología , Demencia/psicología , Forminas , Humanos , Masculino , Memoria/fisiología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas del Tejido Nervioso , Plasticidad Neuronal/genética , Fenotipo , Factores de Riesgo , Trastornos por Estrés Postraumático/complicaciones , Trastornos por Estrés Postraumático/epidemiología , Trastornos por Estrés Postraumático/genética
17.
Proc Natl Acad Sci U S A ; 114(23): E4686-E4694, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28533418

RESUMEN

Schizophrenia is a devastating disease that arises on the background of genetic predisposition and environmental risk factors, such as early life stress (ELS). In this study, we show that ELS-induced schizophrenia-like phenotypes in mice correlate with a widespread increase of histone-deacetylase 1 (Hdac1) expression that is linked to altered DNA methylation. Hdac1 overexpression in neurons of the medial prefrontal cortex, but not in the dorsal or ventral hippocampus, mimics schizophrenia-like phenotypes induced by ELS. Systemic administration of an HDAC inhibitor rescues the detrimental effects of ELS when applied after the manifestation of disease phenotypes. In addition to the hippocampus and prefrontal cortex, mice subjected to ELS exhibit increased Hdac1 expression in blood. Moreover, Hdac1 levels are increased in blood samples from patients with schizophrenia who had encountered ELS, compared with patients without ELS experience. Our data suggest that HDAC1 inhibition should be considered as a therapeutic approach to treat schizophrenia.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Esquizofrenia/enzimología , Estrés Psicológico/enzimología , Adulto , Anciano , Animales , Metilación de ADN , Femenino , Hipocampo/enzimología , Histona Desacetilasa 1/sangre , Histona Desacetilasa 1/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Persona de Mediana Edad , Fenotipo , Corteza Prefrontal/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esquizofrenia/etiología , Esquizofrenia/genética , Estrés Psicológico/complicaciones , Estrés Psicológico/genética , Adulto Joven
18.
Am J Med Genet B Neuropsychiatr Genet ; 174(3): 315-323, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28190298

RESUMEN

Bipolar disorder (BD) is a common, recurring psychiatric illness with unknown pathogenesis. Recent studies suggest that microRNA (miRNA) levels in brains of BD patients are significantly altered, and these changes may offer insight into BD pathology or etiology. Previously, we observed significant alterations of miR-29c levels in extracellular vesicles (EVs) extracted from prefrontal cortex (Brodmann area 9, BA9) of BD patients. In this study, we show that EVs extracted from the anterior cingulate cortex (BA24), a crucial area for modulating emotional expression and affect, have increased levels of miR-149 in BD patients compared to controls. Because miR-149 has been shown to inhibit glial proliferation, increased miR-149 expression in BA24-derived EVs is consistent with the previously reported reduced glial cell numbers in BA24 of patients diagnosed with either familial BD or familial major depressive disorder. qPCR analysis of laser-microdissected neuronal and glial cells from BA24 cortical samples of BD patients verified that the glial, but not neuronal, population exhibits significantly increased miR-149 expression. Finally, we report altered expression of both miR-149 and miR-29c in EVs extracted from brains of Flinders Sensitive Line rats, a well-validated animal model exhibiting depressive-like behaviors and glial (astrocytic) dysfunction. These findings warrant future investigations into the potential of using EV miRNA signatures as biomarkers to further enhance the biological definition of BD. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Trastorno Bipolar/diagnóstico , Trastorno Bipolar/genética , MicroARNs/genética , Animales , Biomarcadores/sangre , Encéfalo/patología , Trastorno Depresivo Mayor/patología , Modelos Animales de Enfermedad , Vesículas Extracelulares/genética , Femenino , Giro del Cíngulo/metabolismo , Humanos , Masculino , MicroARNs/sangre , Ratas
19.
Neuro Oncol ; 19(7): 986-996, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28104717

RESUMEN

BACKGROUND: Clinical genomics platforms are needed to identify targetable alterations, but implementation of these technologies and best practices in routine clinical pediatric oncology practice are not yet well established. METHODS: Profile is an institution-wide prospective clinical research initiative that uses targeted sequencing to identify targetable alterations in tumors. OncoPanel, a multiplexed targeted exome-sequencing platform that includes 300 cancer-causing genes, was used to assess single nucleotide variants and rearrangements/indels. Alterations were annotated (Tiers 1-4) based on clinical significance, with Tier 1 alterations having well-established clinical utility. OncoCopy, a clinical genome-wide array comparative genomic hybridization (aCGH) assay, was also performed to evaluate copy number alterations and better define rearrangement breakpoints. RESULTS: Cancer genomes of 203 pediatric brain tumors were profiled across histological subtypes, including 117 samples analyzed by OncoPanel, 146 by OncoCopy, and 60 tumors subjected to both methodologies. OncoPanel revealed clinically relevant alterations in 56% of patients (44 cancer mutations and 20 rearrangements), including BRAF alterations that directed the use of targeted inhibitors. Rearrangements in MYB-QKI, MYBL1, BRAF, and FGFR1 were also detected. Furthermore, while copy number profiles differed across histologies, the combined use of OncoPanel and OncoCopy identified subgroup-specific alterations in 89% (17/19) of medulloblastomas. CONCLUSION: The combination of OncoPanel and OncoCopy multiplex genomic assays can identify critical diagnostic, prognostic, and treatment-relevant alterations and represents an effective precision medicine approach for clinical evaluation of pediatric brain tumors.


Asunto(s)
Neoplasias Encefálicas/genética , Variaciones en el Número de Copia de ADN , Exoma , Genómica/métodos , Medicina de Precisión/métodos , Neoplasias Encefálicas/diagnóstico , Niño , Hibridación Genómica Comparativa , Dosificación de Gen , Humanos , Mutación , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
20.
Prog Neurobiol ; 151: 101-138, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27084356

RESUMEN

None of the proposed mechanisms of Alzheimer's disease (AD) fully explains the distribution patterns of the neuropathological changes at the cellular and regional levels, and their clinical correlates. One aspect of this problem lies in the complex genetic, epigenetic, and environmental landscape of AD: early-onset AD is often familial with autosomal dominant inheritance, while the vast majority of AD cases are late-onset, with the ε4 variant of the gene encoding apolipoprotein E (APOE) known to confer a 5-20 fold increased risk with partial penetrance. Mechanisms by which genetic variants and environmental factors influence the development of AD pathological changes, especially neurofibrillary degeneration, are not yet known. Here we review current knowledge of the involvement of the monoaminergic systems in AD. The changes in the serotonergic, noradrenergic, dopaminergic, histaminergic, and melatonergic systems in AD are briefly described. We also summarize the possibilities for monoamine-based treatment in AD. Besides neuropathologic AD criteria that include the noradrenergic locus coeruleus (LC), special emphasis is given to the serotonergic dorsal raphe nucleus (DRN). Both of these brainstem nuclei are among the first to be affected by tau protein abnormalities in the course of sporadic AD, causing behavioral and cognitive symptoms of variable severity. The possibility that most of the tangle-bearing neurons of the LC and DRN may release amyloid ß as well as soluble monomeric or oligomeric tau protein trans-synaptically by their diffuse projections to the cerebral cortex emphasizes their selective vulnerability and warrants further investigations of the monoaminergic systems in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Monoaminas Biogénicas/metabolismo , Neuropatología , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Animales , Humanos , Fosforilación , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...