Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 13(1): 16419, 2023 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-37775676

RESUMEN

Major depressive disorder (MDD) and chronic unpredictable stress (CUS) in animals feature comparable cellular and molecular disturbances that involve neurons and glial cells in gray and white matter (WM) in prefrontal brain areas. These same areas demonstrate disturbed connectivity with other brain regions in MDD and stress-related disorders. Functional connectivity ultimately depends on signal propagation along WM myelinated axons, and thus on the integrity of nodes of Ranvier (NRs) and their environment. Various glia-derived proteoglycans interact with NR axonal proteins to sustain NR function. It is unclear whether NR length and the content of associated proteoglycans is altered in prefrontal cortex (PFC) WM of human subjects with MDD and in experimentally stressed animals. The length of WM NRs in histological sections from the PFC of 10 controls and 10 MDD subjects, and from the PFC of control and CUS rats was measured. In addition, in WM of the same brain region, five proteoglycans, tenascin-R and NR protein neurofascin were immunostained or their levels measured with western blots. Analysis of covariance and t-tests were used for group comparisons. There was dramatic reduction of NR length in PFC WM in both MDD and CUS rats. Proteoglycan BRAL1 immunostaining was reduced at NRs and in overall WM of MDD subjects, as was versican in overall WM. Phosphacan immunostaining and levels were increased in both in MDD and CUS. Neurofascin immunostaining at NRs and in overall WM was significantly increased in MDD. Reduced length of NRs and increased phosphacan and neurocan in MDD and stressed animals suggest that morphological and proteoglycan changes at NRs in depression may be related to stress exposure and contribute to connectivity alterations. However, differences between MDD and CUS for some NR related markers may point to other mechanisms affecting the structure and function of NRs in MDD.


Asunto(s)
Trastorno Depresivo Mayor , Sustancia Blanca , Humanos , Ratas , Animales , Sustancia Blanca/patología , Nódulos de Ranvier/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Corteza Prefrontal/metabolismo , Versicanos/metabolismo
2.
Transl Psychiatry ; 12(1): 363, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36064829

RESUMEN

Brodmann Area 46 (BA46) has long been regarded as a hotspot of disease pathology in individuals with schizophrenia (SCH) and major depressive disorder (MDD). Pyramidal neurons in layer III of the Brodmann Area 46 (BA46) project to other cortical regions and play a fundamental role in corticocortical and thalamocortical circuits. The AutoCUTS-LM pipeline was used to study the 3-dimensional structural morphology and spatial organization of pyramidal cells. Using quantitative light microscopy, we used stereology to calculate the entire volume of layer III in BA46 and the total number and density of pyramidal cells. Volume tensors estimated by the planar rotator quantified the volume, shape, and nucleus displacement of pyramidal cells. All of these assessments were carried out in four groups of subjects: controls (C, n = 10), SCH (n = 10), MDD (n = 8), and suicide subjects with a history of depression (SU, n = 11). SCH subjects had a significantly lower somal volume, total number, and density of pyramidal neurons when compared to C and tended to show a volume reduction in layer III of BA46. When comparing MDD subjects with C, the measured parameters were inclined to follow SCH, although there was only a significant reduction in pyramidal total cell number. While no morphometric differences were observed between SU and MDD, SU had a significantly higher total number of pyramidal cells and nucleus displacement than SCH. Finally, no differences in the spatial organization of pyramidal cells were found among groups. These results suggest that despite significant morphological alterations in layer III of BA46, which may impair prefrontal connections in people with SCH and MDD, the spatial organization of pyramidal cells remains the same across the four groups and suggests no defects in neuronal migration. The increased understanding of pyramidal cell biology may provide the cellular basis for symptoms and neuroimaging observations in SCH and MDD patients.


Asunto(s)
Trastorno Depresivo Mayor , Esquizofrenia , Suicidio , Depresión , Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/patología , Humanos , Corteza Prefrontal/patología , Células Piramidales/patología , Esquizofrenia/patología
3.
Commun Biol ; 4(1): 1030, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34475516

RESUMEN

Techniques involving three-dimensional (3D) tissue structure reconstruction and analysis provide a better understanding of changes in molecules and function. We have developed AutoCUTS-LM, an automated system that allows the latest advances in 3D tissue reconstruction and cellular analysis developments using light microscopy on various tissues, including archived tissue. The workflow in this paper involved advanced tissue sampling methods of the human cerebral cortex, an automated serial section collection system, digital tissue library, cell detection using convolution neural network, 3D cell reconstruction, and advanced analysis. Our results demonstrated the detailed structure of pyramidal cells (number, volume, diameter, sphericity and orientation) and their 3D spatial organization are arranged in a columnar structure. The pipeline of these combined techniques provides a detailed analysis of tissues and cells in biology and pathology.


Asunto(s)
Corteza Cerebral/anatomía & histología , Imagenología Tridimensional/métodos , Microtomía , Humanos , Microscopía , Microscopía Electrónica
4.
Genes (Basel) ; 12(8)2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34440415

RESUMEN

Schizophrenia is a genetically complex neuropsychiatric disorder with largely unresolved mechanisms of pathology. Identification of genes and pathways associated with schizophrenia is important for understanding the development, progression and treatment of schizophrenia. In this study, pathways associated with schizophrenia were explored at the level of gene expression. The study included post-mortem brain tissue samples from 68 schizophrenia patients and 44 age and sex-matched control subjects. Whole transcriptome poly-A selected paired-end RNA sequencing was performed on tissue from the prefrontal cortex and orbitofrontal cortex. RNA expression differences were detected between case and control individuals, focusing both on single genes and pathways. The results were validated with RT-qPCR. Significant differential expression between patient and controls groups was found for 71 genes. Gene ontology analysis of differentially expressed genes revealed an up-regulation of multiple genes in immune response among the patients (corrected p-value = 0.004). Several genes in the category belong to the complement system, including C1R, C1S, C7, FCN3, SERPING1, C4A and CFI. The increased complement expression is primarily driven by a subgroup of patients with increased expression of immune/inflammatory response genes, pointing to important differences in disease etiology within the patient group. Weighted gene co-expression network analysis highlighted networks associated with both synaptic transmission and activation of the immune response. Our results demonstrate the importance of immune-related pathways in schizophrenia and provide evidence for elevated expression of the complement cascade as an important pathway in schizophrenia pathology.


Asunto(s)
Proteínas del Sistema Complemento/metabolismo , Perfilación de la Expresión Génica , Cambios Post Mortem , Esquizofrenia/metabolismo , Regulación hacia Arriba , Adulto , Anciano , Femenino , Ontología de Genes , Humanos , Masculino , Persona de Mediana Edad , Esquizofrenia/genética , Esquizofrenia/patología
5.
Transl Psychiatry ; 10(1): 401, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208725

RESUMEN

In recent years, studies have shown higher prevalence of autoantibodies in patients with schizophrenia compared to healthy individuals. This study applies an untargeted and a targeted affinity proteomics approach to explore and characterize the autoantibody repertoire in brain tissues from 73 subjects diagnosed with schizophrenia and 52 control subjects with no psychiatric or neurological disorders. Selected brain tissue lysates were first explored for IgG reactivity on planar microarrays composed of 11,520 protein fragments representing 10,820 unique proteins. Based on these results of ours and other previous studies of autoantibodies related to psychosis, we selected 226 fragments with an average length of 80 amino acids, representing 127 unique proteins. Tissue-based analysis of IgG reactivities using antigen suspension bead arrays was performed in a multiplex and parallel fashion for all 125 subjects. Among the detected autoantigens, higher IgG reactivity in subjects with schizophrenia, as compared to psychiatrically healthy subjects, was found against the glutamate ionotropic receptor NMDA type subunit 2D (anti-GluN2D). In a separate cohort with serum samples from 395 young adults with a wider spectrum of psychiatric disorders, higher levels of serum autoantibodies targeting GluN2D were found when compared to 102 control individuals. By further validating GluN2D and additional potential autoantigens, we will seek insights into how these are associated with severe mental illnesses.


Asunto(s)
Autoanticuerpos , Esquizofrenia , Autoantígenos , Encéfalo , Humanos , Proteómica , Adulto Joven
6.
Geroscience ; 42(5): 1387-1410, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32696219

RESUMEN

Diabetes mellitus (DM) is a leading risk factor for aging-related dementia; however, the underlying mechanisms are not well understood. The present study, utilizing a non-obese T2DN diabetic model, demonstrates that the myogenic response of the middle cerebral artery (MCA) and parenchymal arteriole (PA) and autoregulation of cerebral blood flow (CBF) in the surface and deep cortex were impaired at both young and old ages. The impaired CBF autoregulation was more severe in old than young DM rats, and in the deep than the surface cortex. The myogenic tone of the MCA was enhanced at perfusion pressure in the range of 40-100 mmHg in young DM rats but was reduced at 140-180 mmHg in old DM rats. No change of the myogenic tone of the PA was observed in young DM rats, whereas it was significantly reduced at 30-60 mmHg in old DM rats. Old DM rats had enhanced blood-brain barrier (BBB) leakage and neurodegeneration, reduced vascular density, tight junction, and pericyte coverage on cerebral capillaries in the CA3 region in the hippocampus. Additionally, DM rats displayed impaired functional hyperemia and spatial learning and short- and long-term memory at both young and old ages. Old DM rats had impaired non-spatial short-term memory. These results revealed that impaired CBF autoregulation and enhanced BBB leakage plays an essential role in the pathogenesis of age- and diabetes-related dementia. These findings will lay the foundations for the discovery of anti-diabetic therapies targeting restoring CBF autoregulation to prevent the onset and progression of dementia in elderly DM.


Asunto(s)
Diabetes Mellitus Experimental , Envejecimiento , Animales , Circulación Cerebrovascular , Cognición , Diabetes Mellitus Experimental/complicaciones , Homeostasis , Ratas
7.
J Affect Disord ; 270: 165-173, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32339108

RESUMEN

BACKGROUND: Psychosis is common in bipolar disorder (BD) and is related to more severe cognitive impairments. Since the molecular mechanism of BD psychosis is elusive, we conducted this study to explore the proteomic differences associated with BD psychosis in the dorsolateral prefrontal cortex (DLPFC; BA9). METHODS: Postmortem DLPFC gray matter tissues from five pairs of age-matched male BD subjects with and without psychosis history were used. Tissue proteomes were identified and quantified by label-free liquid chromatography tandem mass spectrometry and then compared between groups. Statistical significance was set at q < 0.40 and Log2 fold change (Log2FC) ≥ |1|. Protein groups with differential expression between groups at p < 0.05 were subjected to pathway analysis. RESULTS: Eleven protein groups differed significantly between groups, including the reduction of tenascin C (q = 0.005, Log2FC = -1.78), the elevations of synaptoporin (q = 0.235, Log2FC = 1.17) and brain-specific angiogenesis inhibitor 1-associated protein 3 (q = 0.241, Log2FC = 2.10) in BD with psychosis. The between-group differences of these proteins were confirmed by Western blots. The top enriched pathways (p < 0.05 with ≥ 3 hits) were the outgrowth of neurons, neuronal cell proliferation, growth of neurites, and outgrowth of neurites, which were all predicted to be upregulated in BD with psychosis. LIMITATIONS: Small sample size and uncertain relationships of the observed proteomic differences with illness stage and acute psychosis. CONCLUSIONS: These results suggested BD with psychosis history may be associated with abnormalities in neurodevelopment, neuroplasticity, neurotransmission, and neuromodulation in the DLPFC.


Asunto(s)
Trastorno Bipolar , Trastornos Psicóticos , Sustancia Gris , Humanos , Imagen por Resonancia Magnética , Masculino , Corteza Prefrontal , Proteómica
8.
J Neurosci ; 40(22): 4418-4431, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32269106

RESUMEN

Emerging evidence suggests that there is a reduction in overall cortical excitatory to inhibitory balance in major depressive disorder (MDD), which afflicts ∼14%-20% of individuals. Reduced pyramidal cell arborization occurs with stress and MDD, and may diminish excitatory neurotransmission. Enhanced deposition of perineuronal net (PNN) components also occurs with stress. Since parvalbumin-expressing interneurons are the predominant cell population that is enveloped by PNNs, which enhance their ability to release GABA, excess PNN deposition likely increases pyramidal cell inhibition. In the present study, we investigate the potential for matrix metalloprotease-9 (MMP-9), an endopeptidase secreted in response to neuronal activity, to contribute to the antidepressant efficacy of the serotonin/norepinephrine reuptake inhibitor venlafaxine in male mice. Chronic venlafaxine increases MMP-9 levels in murine cortex, and increases both pyramidal cell arborization and PSD-95 expression in the cortex of WT but not MMP-9-null mice. We have previously shown that venlafaxine reduces PNN deposition and increases the power of ex vivo γ oscillations in conventionally housed mice. γ power is increased with pyramidal cell disinhibition and with remission from MDD. Herein we observe that PNN expression is increased in a corticosterone-induced stress model of disease and reduced by venlafaxine. Compared with mice that receive concurrent venlafaxine, corticosterone-treated mice also display reduced ex vivo γ power and impaired working memory. Autopsy-derived PFC samples show elevated MMP-9 levels in antidepressant-treated MDD patients compared with controls. These preclinical and postmortem findings highlight a link between extracellular matrix regulation and MDD.SIGNIFICANCE STATEMENT Reduced excitatory neurotransmission occurs with major depressive disorder, and may be normalized by antidepressant treatment. Underlying molecular mechanisms are, however, not well understood. Herein we investigate a potential role for an extracellular protease, released from neurons and known to play a role in learning and memory, in antidepressant-associated increases in excitatory transmission. Our data suggest that this protease, matrix metalloprotease-9, increases branching of excitatory neurons and concomitantly attenuates the perineuronal net to potentially reduce inhibitory input to these neurons. Matrix metalloprotease-9 may thus enhance overall excitatory/inhibitory balance and neuronal population dynamics, which are important to mood and memory.


Asunto(s)
Trastorno Depresivo Mayor/tratamiento farmacológico , Ritmo Gamma , Metaloproteinasa 9 de la Matriz/metabolismo , Inhibición Neural , Inhibidores de Captación de Serotonina y Norepinefrina/farmacología , Estrés Psicológico/complicaciones , Clorhidrato de Venlafaxina/farmacología , Adulto , Anciano , Animales , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Trastorno Depresivo Mayor/etiología , Femenino , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/genética , Memoria a Corto Plazo , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Piramidales/metabolismo , Células Piramidales/patología , Inhibidores de Captación de Serotonina y Norepinefrina/uso terapéutico , Clorhidrato de Venlafaxina/uso terapéutico
9.
Genes (Basel) ; 11(3)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32120974

RESUMEN

Suicide is a major public health concern; nevertheless, its neurobiology remains unknown. An area of interest in suicide research is the dorsolateral prefrontal cortex (DLPFC). We aimed to identify altered proteins and potential biological pathways in the DLPFC of individuals who died by suicide employing mass spectrometry-based untargeted proteomics. Postmortem DLPFC from age-matched male suicide mood disorder cases (n = 5) and non-suicide mood disorder cases (n = 5) were compared. The proteins that differed between groups at false discovery rate (FDR) adjusted p-values (Benjamini-Hochberg-Yekutieli) <0.3 and Log2 fold change (FC) >|0.4| were considered statistically significant and were subjected to pathway analysis by Qiagen Ingenuity software. Thirty-three of the 5162 detected proteins showed significantly altered expression levels in the suicide cases and two of them after adjustment for body mass index. The top differentially expressed protein was potassium voltage-gated channel subfamily Q member 3 (KCNQ3) (Log2FC = -0.481, p = 2.10 × 10-09, FDR = 5.93 × 10-06), which also showed a trend to downregulation in Western blot (p = 0.045, Bonferroni adjusted p = 0.090). The most notably enriched pathway was the GABA receptor signaling pathway (p < 0.001). Here, we report a reduction trend of KCNQ3 levels in the DLPFC of male suicide victims with mood disorders. Further studies with a larger sample size and equal sex representation are needed.


Asunto(s)
Trastornos del Humor/genética , Corteza Prefrontal/metabolismo , Proteínas/genética , Suicidio , Femenino , Humanos , Masculino , Trastornos del Humor/patología , Corteza Prefrontal/patología , Proteómica/tendencias
10.
J Psychiatr Res ; 117: 45-54, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31279243

RESUMEN

Dorsolateral prefrontal cortex (DLPFC) and temporal pole (TP) are brain regions that display abnormalities in bipolar disorder (BD) patients. DNA methylation - an epigenetic mechanism both heritable and sensitive to the environment - may be involved in the pathophysiology of BD. To study BD-associated DNA methylomic differences in these brain regions, we extracted genomic DNA from the postmortem tissues of Brodmann Area (BA) 9 (DLPFC) and BA38 (TP) gray matter from 20 BD, ten major depression (MDD), and ten control age-and-sex-matched subjects. Genome-wide methylation levels were measured using the 850 K Illumina MethylationEPIC BeadChip. We detected striking differences between cortical regions, with greater numbers of between-brain-region differentially methylated positions (DMPs; i.e., CpG sites) in all groups, most pronounced in the BD group, and with substantial overlap across groups. The genes of DMPs common to both BD and MDD (hypothetically associated with their common features such as depression) and those distinct to BD (hypothetically associated with BD-specific features such as mania) were enriched in pathways involved in neurodevelopment including axon guidance. Pathways enriched only in the BD-MDD shared list pointed to GABAergic dysregulation, while those enriched in the BD-only list suggested glutamatergic dysregulation and greater impact on synaptogenesis and synaptic plasticity. We further detected group-specific between-brain-region gene expression differences in ODC1, CALY, GALNT2, and GABRD, which contained significant between-brain-region DMPs. In each brain region, no significant DMPs or differentially methylated regions (DMRs) were found between diagnostic groups. In summary, the methylation differences between DLPFC and TP may provide molecular targets for further investigations of genetic and environmental vulnerabilities associated with both unique and common features of various mood disorders and suggest directions of future development of individualized treatment strategies.


Asunto(s)
Trastorno Bipolar/metabolismo , Metilación de ADN/fisiología , Trastorno Depresivo Mayor/metabolismo , Expresión Génica/fisiología , Genoma/fisiología , Corteza Prefrontal/metabolismo , Lóbulo Temporal/metabolismo , Adulto , Anciano , Autopsia , Islas de CpG , Femenino , Humanos , Masculino , Persona de Mediana Edad
11.
J Psychiatr Res ; 114: 41-47, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31022588

RESUMEN

Schizophrenia is a common mental disorder with high heritability. It is genetically complex and to date more than a hundred risk loci have been identified. Association of environmental factors and schizophrenia has also been reported, while epigenetic analyses have yielded ambiguous and sometimes conflicting results. Here, we analyzed fresh frozen post-mortem brain tissue from a cohort of 73 subjects diagnosed with schizophrenia and 52 control samples, using the Illumina Infinium HumanMethylation450 Bead Chip, to investigate genome-wide DNA methylation patterns in the two groups. Analysis of differential methylation was performed with the Bioconductor Minfi package and modern machine-learning and visualization techniques, which were shown previously to be successful in detecting and highlighting differentially methylated patterns in case-control studies. In this dataset, however, these methods did not uncover any significant signals discerning the patient group and healthy controls, suggesting that if there are methylation changes associated with schizophrenia, they are heterogeneous and complex with small effect.


Asunto(s)
Metilación de ADN/genética , Aprendizaje Automático , Esquizofrenia/genética , Encéfalo/metabolismo , Estudios de Casos y Controles , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Esquizofrenia/metabolismo
12.
JAMA Psychiatry ; 76(6): 634-641, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30840042

RESUMEN

Importance: Monoamine oxidase B (MAO-B) is an important, high-density enzyme in the brain that generates oxidative stress by hydrogen peroxide production, alters mitochondrial function, and metabolizes nonserotonergic monoamines. Recent advances in positron emission tomography radioligand development for MAO-B in humans enable highly quantitative measurement of MAO-B distribution volume (MAO-B VT), an index of MAO-B density. To date, this is the first investigation of MAO-B in the brain of major depressive disorder that evaluates regions beyond the raphe and amygdala. Objective: To investigate whether MAO-B VT is elevated in the prefrontal cortex in major depressive episodes (MDEs) of major depressive disorder. Design, Setting, and Participants: This case-control study was performed at a tertiary care psychiatric hospital from April 1, 2014, to August 30, 2018. Twenty patients with MDEs without current psychiatric comorbidities and 20 age-matched controls underwent carbon 11-labeled [11C]SL25.1188 positron emission tomography scanning to measure MAO-B VT. All participants were drug and medication free, nonsmoking, and otherwise healthy. Main Outcomes and Measures: The MAO-B VT in the prefrontal cortex (PFC). The second main outcome was to evaluate the association between MAO-B VT in the PFC and duration of major depressive disorder illness. Results: Twenty patients with MDEs (mean [SD] age, 34.2 [13.2] years; 11 women) and 20 healthy controls (mean [SD] age, 33.7 [13.1] years; 10 women) were recruited. Patients with MDEs had significantly greater MAO-B VT in the PFC (mean, 26%; analysis of variance, F1,38 = 19.6, P < .001). In individuals with MDEs, duration of illness covaried positively with MAO-B VT in the PFC (analysis of covariance, F1,18 = 15.2, P = .001), as well as most other cortex regions and the thalamus. Conclusions and Relevance: Fifty percent (10 of 20) of patients with MDEs had MAO-B VT values in the PFC exceeding those of healthy controls. Greater MAO-B VT is an index of MAO-B overexpression, which may contribute to pathologies of mitochondrial dysfunction, elevated synthesis of neurotoxic products, and increased metabolism of nonserotonergic monoamines. Hence, this study identifies a common pathological marker associated with downstream consequences poorly targeted by the common selective serotonin reuptake inhibitor treatments. It is also recommended that the highly selective MAO-B inhibitor medications that are compatible for use with other antidepressants and have low risk for hypertensive crisis should be developed or repurposed as adjunctive treatment for MDEs.


Asunto(s)
Trastorno Depresivo Mayor/diagnóstico por imagen , Monoaminooxidasa/metabolismo , Corteza Prefrontal/diagnóstico por imagen , Adulto , Biomarcadores/metabolismo , Radioisótopos de Carbono , Estudios de Casos y Controles , Trastorno Depresivo Mayor/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones , Corteza Prefrontal/metabolismo , Adulto Joven
13.
Methods Mol Biol ; 1938: 247-254, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30617985

RESUMEN

Double immunohistochemistry and confocal microscopy permits to detect the coverage of blood vessels by astrocytic endfeet in human postmortem brain tissue. Here we describe double immunofluorescent staining for detecting a colocalization of a marker of astrocytic processes (labeled with an antibody for aquaporin-4 (AQP4) and a marker for blood vessels (labeled with an antibody to collagen IV). Then, we present a microscopic analysis of the coverage of blood vessels by astrocytic processes using Nikon C1 confocal microscope, Photoshop, and ImageJ software.


Asunto(s)
Astrocitos/metabolismo , Vasos Sanguíneos/metabolismo , Trastorno Depresivo Mayor/etiología , Trastorno Depresivo Mayor/metabolismo , Neuroglía/metabolismo , Astrocitos/patología , Autopsia , Encéfalo/metabolismo , Trastorno Depresivo Mayor/patología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Microscopía Confocal , Neuroglía/patología , Cambios Post Mortem
14.
J Psychiatr Res ; 102: 150-158, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29660602

RESUMEN

Astrocyte functions in white matter are less well understood than in gray matter. Our recent study of white matter in ventral prefrontal cortex (vPFC) revealed alterations in expression of myelin-related genes in major depressive disorder (MDD). Since white matter astrocytes maintain myelin, we hypothesized that morphometry of these cells will be altered in MDD in the same prefrontal white matter region in which myelin-related genes are altered. White matter adjacent to vPFC was examined in 25 MDD and 21 control subjects. Density and size of GFAP-immunoreactive (-ir) astrocyte cell bodies was measured. The area fraction of GFAP-ir astrocytes (cell bodies + processes) was also estimated. GFAP mRNA expression was determined using qRT-PCR. The density of GFAP-ir astrocytes was also measured in vPFC white matter of rats subjected to chronic unpredictable stress (CUS) and control animals. Fibrous and smooth GFAP-ir astrocytes were distinguished in human white matter. The density of both types of astrocytes was significantly decreased in MDD. Area fraction of GFAP immunoreactivity was significantly decreased in MDD, but mean soma size remained unchanged. Expression of GFAP mRNA was significantly decreased in MDD. In CUS rats there was a significant decrease in astrocyte density in prefrontal white matter. The decrease in density and area fraction of white matter astrocytes and GFAP mRNA in MDD may be linked to myelin pathology previously noted in these subjects. Astrocyte pathology may contribute to axon disturbances in axon integrity reported by neuroimaging studies in MDD and interfere with signal conduction in the white matter.


Asunto(s)
Astrocitos/patología , Depresión/patología , Corteza Prefrontal/patología , Sustancia Blanca/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/metabolismo , Depresión/etiología , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Sustancia Blanca/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-29628067

RESUMEN

BACKGROUND: Pathophysiology models of major depression (MD) center on the dysfunction of various cortical areas within the orbital and medial prefrontal cortex. While independent structural and functional abnormalities in these areas are consistent findings in MD, the complex interactions among them and the rest of the cortex remain largely unexplored. METHODS: We used resting-state functional magnetic resonance imaging connectivity to systematically map alterations in the communication between orbital and medial prefrontal cortex fields and the rest of the brain in MD. Functional connectivity (FC) maps from participants with current MD (n = 35), unaffected first-degree relatives (n = 36), and healthy control subjects (n = 38) were subjected to conjunction analyses to distinguish FC markers of MD vulnerability and FC markers of MD disease. RESULTS: FC abnormalities in MD vulnerability were found for dorsal medial wall regions and the anterior insula and concerned altered communication of these areas with the inferior parietal cortex and dorsal posterior cingulate, occipital areas and the brainstem. FC aberrations in current MD included the anterior insula, rostral and dorsal anterior cingulate cortex, and lateral orbitofrontal areas and concerned altered communication with the dorsal striatum, the cerebellum, the precuneus, the anterior prefrontal cortex, somatomotor cortex, dorsolateral prefrontal cortex, and visual areas in the occipital and inferior temporal lobes. CONCLUSIONS: Functionally delineated parcellation maps can be used to identify putative connectivity markers in extended cortical regions such as the orbital and medial prefrontal cortex. The anterior insula and the rostral anterior cingulate cortex play a central role in the pathophysiology of MD, being consistently implicated both in the MD vulnerability and MD disease states.


Asunto(s)
Corteza Cerebral/fisiopatología , Trastorno Depresivo Mayor/fisiopatología , Lóbulo Parietal/fisiopatología , Corteza Prefrontal/fisiopatología , Adulto , Depresión/fisiopatología , Femenino , Giro del Cíngulo/fisiopatología , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Lóbulo Temporal/fisiopatología
16.
Artículo en Inglés | MEDLINE | ID: mdl-30775650

RESUMEN

BACKGROUND: Astrocytes and oligodendrocytes are pathologically altered in dorsolateral prefrontal and orbitofrontal cortices in major depressive disorder. In rat models of stress (major depressive disorder risk factor) astrocyte gap junction protein connexin 43 (Cx43) is reduced in the prelimbic cortex. Astrocyte connexins are recognized to strongly influence myelin maintenance in the central nervous system. However, it is unknown whether stress-related changes in Cx43 and the other major astrocyte connexin, Cx30, occur in the orbitofrontal cortex, or whether connexin changes are concurrent with disturbances in myelination. METHODS: Frozen sections containing prelimbic cortex and orbitofrontal cortex of rats subjected to 35 days of chronic unpredictable stress and controls (n = 6/group) were immunolabeled for Cx43, Cx30, and myelin basic protein. Density of Cx43 or Cx30 immunoreactive puncta and area fraction of myelin basic protein immunoreactivity were measured in prelimbic cortex and orbitofrontal cortex and results analyzed with t test or Pearson correlations. RESULTS: Density of Cx43- and Cx30-positive puncta in both prelimbic cortex and orbitofrontal cortex was lower in chronic unpredictable stress-treated than in control rats. In both regions, the area fraction of myelin basic protein immunoreactivity was also lower in chronic unpredictable stress animals. Myelin basic protein area fraction was positively correlated with the density of Cx43-positive puncta in orbitofrontal cortex, and with Cx30 puncta in prelimbic cortex. CONCLUSION: Low Cx43 and Cx30 after chronic unpredictable stress in rat prelimbic cortex and orbitofrontal cortex suggests that reduced astrocytic gap junction density may generalize to the entire prefrontal cortex. Concurrent reduction of Cx43-, Cx30-, and myelin basic protein-immunolabeled structures is consistent with a mechanism linking changes in astrocyte gap junction proteins and disturbed myelin morphology in depression.

17.
Cereb Cortex ; 28(9): 3129-3142, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28968778

RESUMEN

Molecular mechanisms that define patterns of neuropeptide expression are essential for the formation and rewiring of neural circuits. The prodynorphin gene (PDYN) gives rise to dynorphin opioid peptides mediating depression and substance dependence. We here demonstrated that PDYN is expressed in neurons in human dorsolateral prefrontal cortex (dlPFC), and identified neuronal differentially methylated region in PDYN locus framed by CCCTC-binding factor binding sites. A short, nucleosome size human-specific promoter CpG island (CGI), a core of this region may serve as a regulatory module, which is hypomethylated in neurons, enriched in 5-hydroxymethylcytosine, and targeted by USF2, a methylation-sensitive E-box transcription factor (TF). USF2 activates PDYN transcription in model systems, and binds to nonmethylated CGI in dlPFC. USF2 and PDYN expression is correlated, and USF2 and PDYN proteins are co-localized in dlPFC. Segregation of activatory TF and repressive CGI methylation may ensure contrasting PDYN expression in neurons and glia in human brain.


Asunto(s)
Encefalinas/biosíntesis , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Precursores de Proteínas/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Metilación de ADN/genética , Encefalinas/genética , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Precursores de Proteínas/genética , Transcripción Genética , Factores Estimuladores hacia 5'/metabolismo
18.
Neuroscience ; 359: 30-39, 2017 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-28711621

RESUMEN

Studies of major depressive disorder (MDD) in postmortem brain tissue report enhanced binding to inhibitory serotonin-1A autoreceptors in midbrain dorsal raphe and reductions in length of axons expressing the serotonin transporter (SERT) in dorsolateral prefrontal cortex. The length density of axons expressing SERT in the orbitofrontal cortex (OFC) was determined in 18 subjects with MDD and 17 age-matched control subjects. A monoclonal antibody was used to immunohistochemically label the SERT in fixed sections of OFC. The 3-dimensional length density of SERT-immunoreactive (ir) axons in layer VI of OFC was estimated. The age of subjects with MDD was negatively correlated with SERT axon length (r=-0.77, p<0.0005). The significant effect of age persisted when removing four depressed subjects with an antidepressant medication present at the time of death, or when removing nine depressed subjects that had a recent prescription for an antidepressant medication. Neither gender, tissue pH, postmortem interval, 5-HTTLPR genotype, time in fixative, nor death by suicide had a significant effect on axon length. The age-related decrease in SERT-ir axon length in MDD may reflect pathology of ascending axons passing through deep white matter hyperintensities. Greater length of axons expressing SERT in younger subjects with MDD may result in a significant deficit in serotonin availability in OFC.


Asunto(s)
Axones/metabolismo , Axones/patología , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/patología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad
19.
Brain Struct Funct ; 222(7): 2941-2960, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28255676

RESUMEN

The orbital and medial prefrontal cortex (OMPFC) has been implicated in decision-making, reward and emotion processing, and psychopathology, such as depression and obsessive-compulsive disorder. Human and monkey anatomical studies indicate the presence of various cortical subdivisions and suggest that these are organized in two extended networks, a medial and an orbital one. Attempts have been made to replicate these neuroanatomical findings in vivo using MRI techniques for imaging connectivity. These revealed several consistencies, but also many inconsistencies between reported results. Here, we use fMRI resting-state functional connectivity (FC) and data-driven modularity optimization to parcellate the OMPFC to investigate replicability of in vivo parcellation more systematically. By collecting two resting-state data sets per participant, we were able to quantify the reliability of the observed modules and their boundaries. Results show that there was significantly more than chance overlap in modules and their boundaries at the level of individual data sets. Moreover, some of these consistent boundaries significantly co-localized across participants. Hierarchical clustering showed that the whole-brain FC profiles of the OMPFC subregions separate them in two networks, a medial and orbital one, which overlap with the organization proposed by Barbas and Pandya (J Comp Neurol 286:353-375, 1989) and Ongür and Price (Cereb Cortex 10:206-219, 2000). We conclude that in vivo resting-state FC can delineate reliable and neuroanatomically plausible subdivisions that agree with established cytoarchitectonic trends and connectivity patterns, while other subdivisions do not show the same consistency across data sets and studies.


Asunto(s)
Mapeo Encefálico , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/fisiología , Corteza Prefrontal/diagnóstico por imagen , Adolescente , Adulto , Análisis por Conglomerados , Femenino , Lateralidad Funcional/fisiología , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Oxígeno/sangre , Descanso , Adulto Joven
20.
Biochim Biophys Acta Gen Subj ; 1861(2): 246-255, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27838394

RESUMEN

BACKGROUND: Neuropeptide precursors are traditionally viewed as proteins giving rise to small neuropeptide molecules. Prodynorphin (PDYN) is the precursor protein to dynorphins, endogenous ligands for the κ-opioid receptor. Alternative mRNA splicing of neuropeptide genes may regulate cell- and tissue-specific neuropeptide expression and produce novel protein isoforms. We here searched for novel PDYN mRNA and their protein product in the human brain. METHODS: Novel PDYN transcripts were identified using nested PCR amplification of oligo(dT) selected full-length capped mRNA. Gene expression was analyzed by qRT-PCR, PDYN protein by western blotting and confocal imaging, dynorphin peptides by radioimmunoassay. Neuronal nuclei were isolated using fluorescence-activated nuclei sorting (FANS) from postmortem human striatal tissue. Immunofluorescence staining and confocal microscopy was performed for human caudate nucleus. RESULTS: Two novel human PDYN mRNA splicing variants were identified. Expression of one of them was confined to the striatum where its levels constituted up to 30% of total PDYN mRNA. This transcript may be translated into ∆SP-PDYN protein lacking 13 N-terminal amino acids, a fragment of signal peptide (SP). ∆SP-PDYN was not processed to mature dynorphins and surprisingly, was targeted to the cell nuclei in a model cellular system. The endogenous PDYN protein was identified in the cell nuclei in human striatum by western blotting of isolated neuronal nuclei, and by confocal imaging. CONCLUSIONS AND GENERAL SIGNIFICANCE: High levels of alternatively spliced ∆SP-PDYN mRNA and nuclear localization of PDYN protein suggests a nuclear function for this isoform of the opioid peptide precursor in human striatum.


Asunto(s)
Núcleo Caudado/metabolismo , Núcleo Celular/metabolismo , Péptidos Opioides/metabolismo , Isoformas de Proteínas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Aminoácidos/metabolismo , Animales , Línea Celular Tumoral , Dinorfinas/metabolismo , Encefalinas/metabolismo , Femenino , Regulación de la Expresión Génica/fisiología , Silenciador del Gen/fisiología , Humanos , Masculino , Persona de Mediana Edad , Precursores de Proteínas/metabolismo , ARN Mensajero/metabolismo , Ratas , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...