Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
iScience ; 27(1): 108725, 2024 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-38226160

RESUMEN

Sorting receptor SORCS2 is a stress-response factor protecting neurons from acute insults, such as during epilepsy. SORCS2 is also expressed in the pancreas, yet its action in this tissue remains unknown. Combining metabolic studies in SORCS2-deficient mice with ex vivo functional analyses and single-cell transcriptomics of pancreatic tissues, we identified a role for SORCS2 in protective stress response in pancreatic islets, essential to sustain insulin release. We show that SORCS2 is predominantly expressed in islet alpha cells. Loss of expression coincides with inability of these cells to produce osteopontin, a secreted factor that facilitates insulin release from stressed beta cells. In line with diminished osteopontin levels, beta cells in SORCS2-deficient islets show gene expression patterns indicative of aggravated cell stress, and exhibit defects in insulin granule maturation and a blunted glucose response. These findings corroborate a function for SORCS2 in protective stress response that extends to metabolism.

2.
Nat Commun ; 14(1): 7871, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052784

RESUMEN

Current differentiation protocols for generating mesencephalic dopaminergic (mesDA) neurons from human pluripotent stem cells result in grafts containing only a small proportion of mesDA neurons when transplanted in vivo. In this study, we develop lineage-restricted undifferentiated stem cells (LR-USCs) from pluripotent stem cells, which enhances their potential for differentiating into caudal midbrain floor plate progenitors and mesDA neurons. Using a ventral midbrain protocol, 69% of LR-USCs become bona fide caudal midbrain floor plate progenitors, compared to only 25% of human embryonic stem cells (hESCs). Importantly, LR-USCs generate significantly more mesDA neurons under midbrain and hindbrain conditions in vitro and in vivo. We demonstrate that midbrain-patterned LR-USC progenitors transplanted into 6-hydroxydopamine-lesioned rats restore function in a clinically relevant non-pharmacological behavioral test, whereas midbrain-patterned hESC-derived progenitors do not. This strategy demonstrates how lineage restriction can prevent the development of undesirable lineages and enhance the conditions necessary for mesDA neuron generation.


Asunto(s)
Neuronas Dopaminérgicas , Células Madre Pluripotentes , Humanos , Ratas , Animales , Neuronas Dopaminérgicas/metabolismo , Factores de Transcripción/metabolismo , Diferenciación Celular/fisiología , Mesencéfalo , Células Madre Pluripotentes/metabolismo
3.
Cell Rep ; 42(11): 113333, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37897724

RESUMEN

Motor neuron (MN) development and nerve regeneration requires orchestrated action of a vast number of molecules. Here, we identify SorCS2 as a progranulin (PGRN) receptor that is required for MN diversification and axon outgrowth in zebrafish and mice. In zebrafish, SorCS2 knockdown also affects neuromuscular junction morphology and fish motility. In mice, SorCS2 and PGRN are co-expressed by newborn MNs from embryonic day 9.5 until adulthood. Using cell-fate tracing and nerve segmentation, we find that SorCS2 deficiency perturbs cell-fate decisions of brachial MNs accompanied by innervation deficits of posterior nerves. Additionally, adult SorCS2 knockout mice display slower motor nerve regeneration. Interestingly, primitive macrophages express high levels of PGRN, and their interaction with SorCS2-positive motor axon is required during axon pathfinding. We further show that SorCS2 binds PGRN to control its secretion, signaling, and conversion into granulins. We propose that PGRN-SorCS2 signaling controls MN development and regeneration in vertebrates.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Pez Cebra , Ratones , Animales , Progranulinas , Pez Cebra/metabolismo , Neuronas Motoras/metabolismo , Granulinas , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo
4.
Nat Med ; 29(7): 1832-1844, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37464041

RESUMEN

Depression is a common psychiatric disorder and a leading cause of disability worldwide. Here we conducted a genome-wide association study meta-analysis of six datasets, including >1.3 million individuals (371,184 with depression) and identified 243 risk loci. Overall, 64 loci were new, including genes encoding glutamate and GABA receptors, which are targets for antidepressant drugs. Intersection with functional genomics data prioritized likely causal genes and revealed new enrichment of prenatal GABAergic neurons, astrocytes and oligodendrocyte lineages. We found depression to be highly polygenic, with ~11,700 variants explaining 90% of the single-nucleotide polymorphism heritability, estimating that >95% of risk variants for other psychiatric disorders (anxiety, schizophrenia, bipolar disorder and attention deficit hyperactivity disorder) were influencing depression risk when both concordant and discordant variants were considered, and nearly all depression risk variants influenced educational attainment. Additionally, depression genetic risk was associated with impaired complex cognition domains. We dissected the genetic and clinical heterogeneity, revealing distinct polygenic architectures across subgroups of depression and demonstrating significantly increased absolute risks for recurrence and psychiatric comorbidity among cases of depression with the highest polygenic burden, with considerable sex differences. The risks were up to 5- and 32-fold higher than cases with the lowest polygenic burden and the background population, respectively. These results deepen the understanding of the biology underlying depression, its disease progression and inform precision medicine approaches to treatment.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno Bipolar , Esquizofrenia , Masculino , Femenino , Humanos , Estudio de Asociación del Genoma Completo , Depresión , Trastorno Bipolar/epidemiología , Trastorno Bipolar/genética , Esquizofrenia/epidemiología , Esquizofrenia/genética , Trastorno por Déficit de Atención con Hiperactividad/epidemiología , Trastorno por Déficit de Atención con Hiperactividad/genética , Polimorfismo de Nucleótido Simple/genética , Predisposición Genética a la Enfermedad
5.
Transl Psychiatry ; 12(1): 319, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35941107

RESUMEN

Bromodomain containing 1 (BRD1) encodes an epigenetic regulator that controls the expression of genetic networks linked to mental illness. BRD1 is essential for normal brain development and its role in psychopathology has been demonstrated in genetic and preclinical studies. However, the neurobiology that bridges its molecular and neuropathological effects remains poorly explored. Here, using publicly available datasets, we find that BRD1 targets nuclear genes encoding mitochondrial proteins in cell lines and that modulation of BRD1 expression, irrespective of whether it is downregulation or upregulation of one or the other existing BRD1 isoforms (BRD1-L and BRD1-S), leads to distinct shifts in the expression profile of these genes. We further show that the expression of nuclear genes encoding mitochondrial proteins is negatively correlated with the expression of BRD1 mRNA during human brain development. In accordance, we identify the key gate-keeper of mitochondrial metabolism, Peroxisome proliferator-activated receptor (PPAR) among BRD1's co-transcription factors and provide evidence that BRD1 acts as a co-repressor of PPAR-mediated transcription. Lastly, when using quantitative PCR, mitochondria-targeted fluorescent probes, and the Seahorse XFe96 Analyzer, we demonstrate that modulation of BRD1 expression in cell lines alters mitochondrial physiology (mtDNA content and mitochondrial mass), metabolism (reducing power), and bioenergetics (among others, basal, maximal, and spare respiration) in an expression level- and isoform-dependent manner. Collectively, our data suggest that BRD1 is a transcriptional regulator of nuclear-encoded mitochondrial proteins and that disruption of BRD1's genomic actions alters mitochondrial functions. This may be the mechanism underlying the cellular and atrophic changes of neurons previously associated with BRD1 deficiency and suggests that mitochondrial dysfunction may be a possible link between genetic variation in BRD1 and psychopathology in humans.


Asunto(s)
Histona Acetiltransferasas , Esquizofrenia , Metabolismo Energético , Histona Acetiltransferasas/fisiología , Humanos , Mitocondrias/metabolismo , Proteínas Mitocondriales , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Isoformas de Proteínas/metabolismo , Esquizofrenia/genética
6.
Genes (Basel) ; 13(6)2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35741801

RESUMEN

Bipolar disorder is a debilitating psychiatric condition that is shaped in a concerted interplay between hereditary and triggering risk factors. Profound depression and mania define the disorder, but high clinical heterogeneity among patients complicates diagnosis as well as pharmacological intervention. Identification of peripheral biomarkers that capture the genomic response to the exposome may thus progress the development of personalized treatment. MicroRNAs (miRNAs) play a prominent role in of post-transcriptional gene regulation in the context of brain development and mental health. They are coordinately modulated by multifarious effectors, and alteration in their expression profile has been reported in a variety of psychiatric conditions. Intriguingly, miRNAs can be released from CNS cells and enter circulatory bio-fluids where they remain remarkably stable. Hence, peripheral circulatory miRNAs may act as bio-indicators for the combination of genetic risk, environmental exposure, and/or treatment response. Here we provide a comprehensive literature search and data mining approach that summarize current experimental evidence supporting the applicability of miRNAs for patient stratification in bipolar disorder.


Asunto(s)
Trastorno Bipolar , MicroARN Circulante , MicroARNs , Biomarcadores , Trastorno Bipolar/diagnóstico , Trastorno Bipolar/genética , MicroARN Circulante/genética , Minería de Datos , Humanos , MicroARNs/genética , MicroARNs/metabolismo
7.
Cytometry A ; 101(4): 298-310, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34842347

RESUMEN

Isolation of multiple cell populations from limited starting material and with minimal influence on cell homeostasis and viability are common requirements in both basic and clinical research. Fluorescence-activated cell sorting (FACS) is the most commonly applied sorting methodology with the majority of instruments being based on high pressure and electrostatic deflection. A more recent technology is based on a mechanical valve, operating at low pressure. In the present work we compared the two technologies by parallel sorting of small amounts of peripheral blood and umbilical cord blood on a BD FACSAria™ III and Miltenyi MACSQuant® Tyto® instrument. Concurrent manually performed magnetic-based cell sorting served as reference. Sorting metrics, including purity and viability, were compared. Expression of the heat-shock protein HSPA1A immediately post sorting and the proliferation potential of sorted T-cells in vitro was assessed. In general, there was little to distinguish the two fluorescence-activated technologies with regard to sorting metrics and HSPA1A expression. Variation, however, with respect to recovery and viability, was much smaller among Tyto sorted samples. The proliferation potential of Tyto-sorted T-cells was significantly higher compared to Aria-sorted T-cells, indicating that T-cells of the Tyto instrument are less perturbed. In summary, cell types of blood origin including CD34+ cells could effectively be isolated from small input amounts with either fluorescence-activated technology with little immediate effect on viability. The mechanical valve-based sorting by the Tyto instrument; however, appeared to perturb the cells to a lesser extent as judged by their proliferation potential.


Asunto(s)
Sangre Fetal , Separación Celular/métodos , Citometría de Flujo/métodos , Electricidad Estática
8.
J Cell Biol ; 220(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34779857

RESUMEN

Visceral adipose tissue shows remarkable plasticity, constantly replacing mature adipocytes from an inherent pool of adipocyte precursors. The number of precursors is set in the juvenile organism and remains constant in adult life. Which signals drive precursor pool expansion in juveniles and why they operate in visceral but not in subcutaneous white adipose tissue (WAT) are unclear. Using mouse models, we identified the insulin-sensitizing receptor SORLA as a molecular factor explaining the distinct proliferative capacity of visceral WAT. High levels of SORLA activity in precursors of juvenile visceral WAT prime these cells for nutritional stimuli provided through insulin, promoting mitotic expansion of the visceral precursor cell pool in overfed juvenile mice. SORLA activity is low in subcutaneous precursors, blunting their response to insulin and preventing diet-induced proliferation of this cell type. Our findings provide a molecular explanation for the unique proliferative properties of juvenile visceral WAT, and for the genetic association of SORLA with visceral obesity in humans.


Asunto(s)
Adipocitos/citología , Insulina/farmacología , Grasa Intraabdominal/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Receptores de LDL/metabolismo , Células Madre/citología , Células Madre/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Índice de Masa Corporal , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitógenos/farmacología , Células Madre/efectos de los fármacos , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/metabolismo , Adulto Joven
9.
Clin Epigenetics ; 13(1): 200, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34715912

RESUMEN

BACKGROUND: Depression is a common, complex, and debilitating mental disorder estimated to be under-diagnosed and insufficiently treated in society. Liability to depression is influenced by both genetic and environmental risk factors, which are both capable of impacting DNA methylation (DNAm). Accordingly, numerous studies have researched for DNAm signatures of this disorder. Recently, an epigenome-wide association study of monozygotic twins identified an association between DNAm status in the KLK8 (neuropsin) promoter region and severity of depression symptomatology. METHODS: In this study, we aimed to investigate: (i) if blood DNAm levels, quantified by pyrosequencing, at two CpG sites in the KLK8 promoter are associated with depression symptomatology and depression diagnosis in an independent clinical cohort and (ii) if KLK8 DNAm levels are associated with depression, postpartum depression, and depression symptomatology in four independent methylomic cohorts, with blood and brain DNAm quantified by either MBD-seq or 450 k methylation array. RESULTS: DNAm levels in KLK8 were not significantly different between depression cases and controls, and were not significantly associated with any of the depression symptomatology scores after correction for multiple testing (minimum p value for KLK8 CpG1 = 0.12 for 'Depressed mood,' and for CpG2 = 0.03 for 'Loss of self-confidence with other people'). However, investigation of the link between KLK8 promoter DNAm levels and depression-related phenotypes collected from four methylomic cohorts identified significant association (p value < 0.05) between severity of depression symptomatology and blood DNAm levels at seven CpG sites. CONCLUSIONS: Our findings suggest that variance in blood DNAm levels in KLK8 promoter region is associated with severity of depression symptoms, but not depression diagnosis.


Asunto(s)
Metilación de ADN/genética , Depresión/diagnóstico , Calicreínas/análisis , Calicreínas/genética , Anciano , Depresión/psicología , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Humanos , Masculino , Persona de Mediana Edad
10.
PLoS One ; 16(6): e0252799, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34086837

RESUMEN

AIMS: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) binds to angiotensin converting enzyme 2 (ACE2) enabling entrance of the virus into cells and causing the infection termed coronavirus disease of 2019 (COVID-19). Here, we investigate associations between plasma ACE2 and outcome of COVID-19. METHODS AND RESULTS: This analysis used data from a large longitudinal study of 306 COVID-19 positive patients and 78 COVID-19 negative patients (MGH Emergency Department COVID-19 Cohort). Comprehensive clinical data were collected on this cohort, including 28-day outcomes. The samples were run on the Olink® Explore 1536 platform which includes measurement of the ACE2 protein. High admission plasma ACE2 in COVID-19 patients was associated with increased maximal illness severity within 28 days with OR = 1.8, 95%-CI: 1.4-2.3 (P < 0.0001). Plasma ACE2 was significantly higher in COVID-19 patients with hypertension compared with patients without hypertension (P = 0.0045). Circulating ACE2 was also significantly higher in COVID-19 patients with pre-existing heart conditions and kidney disease compared with patients without these pre-existing conditions (P = 0.0363 and P = 0.0303, respectively). CONCLUSION: This study suggests that measuring plasma ACE2 is potentially valuable in predicting COVID-19 outcomes. Further, ACE2 could be a link between COVID-19 illness severity and its established risk factors hypertension, pre-existing heart disease and pre-existing kidney disease.


Asunto(s)
Enzima Convertidora de Angiotensina 2/sangre , COVID-19 , Cardiopatías , Hospitalización , Enfermedades Renales , SARS-CoV-2/metabolismo , Adolescente , Adulto , COVID-19/sangre , COVID-19/mortalidad , COVID-19/terapia , Comorbilidad , Femenino , Cardiopatías/sangre , Cardiopatías/mortalidad , Cardiopatías/terapia , Humanos , Enfermedades Renales/sangre , Enfermedades Renales/mortalidad , Enfermedades Renales/terapia , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad
11.
Mol Neurobiol ; 58(9): 4495-4505, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34056693

RESUMEN

Genetic studies have repeatedly shown that the Bromodomain containing 1 gene, BRD1, is involved in determining mental health, and the importance of the BRD1 protein for normal brain function has been studied in both cell models and constitutive haploinsufficient Brd1+/- mice. Homozygosity for inactivated Brd1 alleles is lethal during embryonic development in mice. In order to further characterize the molecular functions of BRD1 in the brain, we have developed a novel Brd1 knockout mouse model (Brd1-/-) with bi-allelic conditional inactivation of Brd1 in the central nervous system. Brd1-/- mice were viable but smaller and with reduced muscle strength. They showed reduced exploratory behavior and increased sensitivity to pentylenetetrazole-induced seizures supporting the previously described GABAergic dysfunction in constitutive Brd1+/- mice. Because BRD1 takes part in protein complexes with histone binding and modifying functions, we investigated the effect of BRD1 depletion on the global histone modification pattern in mouse brain by mass spectrometry. We found decreased levels of histone H3 acetylation (H3K9ac, H3K14ac, and H3K18ac) and increased N-tail clipping in consequence of BRD1 depletion. Collectively, the presented results support that BRD1 controls gene expression at the epigenetic level by regulating histone H3 proteoforms in the brain.


Asunto(s)
Encéfalo/metabolismo , Histona Acetiltransferasas/genética , Histonas/metabolismo , Esquizofrenia/genética , Convulsiones/genética , Acetilación , Animales , Histona Acetiltransferasas/metabolismo , Histonas/genética , Ratones , Ratones Noqueados , Esquizofrenia/metabolismo , Convulsiones/metabolismo
12.
Mol Neurobiol ; 58(5): 2075-2087, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33415685

RESUMEN

Neurons produced by reprogramming of other cell types are used to study cellular mechanisms of age-related neurodegenerative diseases. To model Alzheimer's disease and other tauopathies, it is essential that alternative splicing of the MAPT transcript in these neurons produces the relevant tau isoforms. Human neurons derived from induced pluripotent stem cells, however, express tau isoform compositions characteristic of foetal neurons rather than of adult neurons unless cultured in vitro for extended time periods. In this study, we characterised the dynamics of the MAPT and APP alternative splicing during a developmental time-course of porcine and murine cerebral cortices. We found age-dependent and species-specific isoform composition of MAPT, including 3R and 4R isoforms in the porcine adult brain similar to that of the adult human brain. We converted adult and embryonic fibroblasts directly into induced neurons and found similar developmental patterns of isoform composition, notably, the 3R and 4R isoforms relevant to the pathogenesis of Alzheimer's disease. Also, we observed cell-type-specific isoform expression of APP transcripts during the conversion. The approach was further used to generate induced neurons from transgenic pigs carrying Alzheimer's disease-causing mutations. We show that such neurons authentically model the first crucial steps in AD pathogenesis.


Asunto(s)
Envejecimiento/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Proteínas tau/metabolismo , Empalme Alternativo , Animales , Animales Modificados Genéticamente , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Porcinos
13.
Cereb Cortex ; 31(3): 1609-1621, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33123721

RESUMEN

The 15q13.3 deletion is associated with multiple neurodevelopmental disorders including epilepsy, schizophrenia, and autism. The Df(h15q13)/+ mouse model was recently generated that recapitulates several phenotypic features of the human 15q13.3 deletion syndrome (DS). However, the biological substrates underlying these phenotypes in Df(h15q13)/+ mice have not yet been fully characterized. RNA sequencing followed by real-time quantitative PCR, western blotting, liquid chromatography-mass spectrometry, and stereological analysis were employed to dissect the molecular, structural, and neurochemical phenotypes of the medial prefrontal cortex (mPFC) circuits in Df(h15q13)/+ mouse model. Transcriptomic profiling revealed enrichment for astrocyte-specific genes among differentially expressed genes, translated by a decrease in the number of glial fibrillary acidic protein positive cells in mPFC of Df(h15q13)/+ mice compared with wild-type mice. mPFC in Df(h15q13)/+ mice also showed a deficit of the inhibitory presynaptic marker GAD65, in addition to a reduction in dendritic arborization and spine density of pyramidal neurons from layers II/III. mPFC levels of GABA and glutamate neurotransmitters were not different between genotypes. Our results suggest that the 15q13.3 deletion modulates nonneuronal circuits in mPFC and confers molecular and morphometric alterations in the inhibitory and excitatory neurocircuits, respectively. These alterations potentially contribute to the phenotypes accompanied with the 15q13.3DS.


Asunto(s)
Astrocitos/patología , Trastornos de los Cromosomas/patología , Trastornos de los Cromosomas/fisiopatología , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Corteza Prefrontal/patología , Corteza Prefrontal/fisiopatología , Convulsiones/patología , Convulsiones/fisiopatología , Sinapsis/patología , Animales , Deleción Cromosómica , Cromosomas Humanos Par 15 , Modelos Animales de Enfermedad , Masculino , Ratones
14.
Stem Cell Res ; 48: 101984, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32971463

RESUMEN

Direct neuronal conversion describes the process of generating induced neurons from somatic cells such as fibroblasts by overexpressing cell type-specific transcription factors, microRNAs or by culturing in the presence of small molecules. This was first achieved by expressing Brn2, Ascl1 and Myt1L in mouse fibroblasts, and was later achieved in human cells by the inclusion of additional factors such as NeuroD1. Here, we present the first protocol for directly converting porcine fibroblasts into induced neurons. We used lentivirus-mediated delivery of previously identified neuron-specifying transcription factors and microRNAs and evaluated morphology and neuron marker expression after ten days of conversion. We found that Ascl1 and microRNAs, miR-9/9* and miR-124 together generated more neuronal cells than other conditions tested. The porcine induced neurons expressed common mature markers such as MAP2 and Synaptophysin after four weeks of conversion. Transcriptomic analysis revealed that fibroblast-specific signatures were silenced early in the conversion process, while the neuron-specific genes became more abundant during conversion. We generated a heterogeneous population of glutamatergic and GABAergic neurons.


Asunto(s)
Reprogramación Celular , MicroARNs , Animales , Fibroblastos , MicroARNs/genética , Neuronas , Porcinos , Factores de Transcripción/genética
15.
Mol Neurobiol ; 57(12): 4978-4988, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32820460

RESUMEN

The 22q11.2 deletion has been identified as a risk factor for multiple neurodevelopmental disorders. Behavioral and cognitive impairments are common among carriers of the 22q11.2 deletion. Parvalbumin expressing (PV+) interneurons provide perisomatic inhibition of excitatory neuronal circuits through GABAA receptors, and a deficit of PV+ inhibitory circuits may underlie a multitude of the behavioral and functional deficits in the 22q11.2 deletion syndrome. We investigated putative deficits of PV+ inhibitory circuits and the associated molecular, morphological, and functional alterations in the prefrontal cortex (PFC) of the Df(h22q11)/+ mouse model of the 22q11.2 hemizygous deletion. We detected a significant decrease in the number of PV+ interneurons in layers II/III of PFC in Df(h22q11)/+ mice together with a reduction in the mRNA and protein levels of GABAA (α3), a PV+ putative postsynaptic receptor subunit. Pyramidal neurons from the same layers further experienced morphological reorganizations of spines and dendrites. Accordingly, a decrease in the levels of the postsynaptic density protein 95 (PSD95) and a higher neuronal activity in response to the GABAA antagonist bicuculline were measured in these layers in PFC of Df(h22q11)/+ mice compared with their wild-type littermates. Our study shows that a hemizygotic deletion of the 22q11.2 locus leads to deficit in the GABAergic control of network activity and involves molecular and morphological changes in both the inhibitory and excitatory synapses of parvalbumin interneurons and pyramidal neurons specifically in layers II/III PFC.


Asunto(s)
Forma de la Célula , Síndrome de DiGeorge/patología , Interneuronas/patología , Parvalbúminas/metabolismo , Corteza Prefrontal/patología , Animales , Bicuculina/farmacología , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large/metabolismo , Antagonistas de Receptores de GABA-A/farmacología , Interneuronas/metabolismo , Masculino , Ratones , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células Piramidales/metabolismo , Células Piramidales/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
16.
Transl Psychiatry ; 10(1): 239, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32681022

RESUMEN

The schizophrenia-associated gene, BRD1, encodes an epigenetic regulator in which chromatin interactome is enriched with genes implicated in mental health. Alterations in histone modifications and epigenetic regulation contribute to brain transcriptomic changes in affective disorders and preclinical data supports a role for BRD1 in psychopathology. However, the implication of BRD1 on affective pathology remains poorly understood. In this study, we assess affective behaviors and associated neurobiology in Brd1+/- mice along with their responses to Fluoxetine and Imipramine. This involves behavioral, neurostructural, and neurochemical characterizations along with regional cerebral gene expression profiling combined with integrative functional genomic analyses. We report behavioral changes in female Brd1+/- mice with translational value to depressive symptomatology that can be alleviated by the administration of antidepressant medications. Behavioral changes are accompanied by altered brain morphometry and imbalances in monoaminergic systems. In accordance, gene expression changes across brain tissues reveal altered neurotransmitter signaling and cluster in functional pathways associated with depression including 'Adrenergic-, GPCR-, cAMP-, and CREB/CREM-signaling'. Integrative gene expression analysis specifically links changes in amygdaloid intracellular signaling activity to the behavioral treatment response in Brd1+/- mice. Collectively, our study highlights the importance of BRD1 as a modulator of affective pathology and adds to our understanding of the molecular mechanisms underlying affective disorders and their treatment response.


Asunto(s)
Histona Acetiltransferasas , Esquizofrenia , Animales , Depresión/genética , Epigénesis Genética , Femenino , Expresión Génica , Ratones , Esquizofrenia/genética
17.
J Physiol Sci ; 70(1): 26, 2020 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-32414324

RESUMEN

Neuropsin is a brain-expressed extracellular matrix serine protease that governs synaptic plasticity through activity-induced proteolytic cleavage of synaptic proteins. Its substrates comprise several molecules central to structural synaptic plasticity, and studies in rodents have documented its role in cognition and the behavioral and neurobiological response to stress. Intriguingly, differential usage of KLK8 (neuropsin gene) splice forms in the fetal and adult brain has only been reported in humans, suggesting that neuropsin may serve a specialized role in human neurodevelopment. Through systematic interrogation of large-scale genetic data, we review KLK8 regulation in the context of mental health and provide a summary of clinical and preclinical evidence supporting a role for neuropsin in the pathogenesis of mental illness.


Asunto(s)
Calicreínas/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos Mentales/metabolismo , Opsinas/metabolismo , Animales , Humanos , Trastornos Mentales/fisiopatología , Salud Mental , Plasticidad Neuronal
18.
J Immunol ; 204(5): 1345-1361, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31969389

RESUMEN

Aggregation of α-synuclein (αSN) is an important histological feature of Parkinson disease. Recent studies showed that the release of misfolded αSN from human and rodent neurons is relevant to the progression and spread of αSN pathology. Little is known, however, about the mechanisms responsible for clearance of extracellular αSN. This study found that human complement receptor (CR) 4 selectively bound fibrillar αSN, but not monomeric species. αSN is an abundant protein in the CNS, which potentially could overwhelm clearance of cytotoxic αSN species. The selectivity of CR4 toward binding fibrillar αSN consequently adds an important αSN receptor function for maintenance of brain homeostasis. Based on the recently solved structures of αSN fibrils and the known ligand preference of CR4, we hypothesize that the parallel monomer stacking in fibrillar αSN creates a known danger-associated molecular pattern of stretches of anionic side chains strongly bound by CR4. Conformational change in the receptor regulated tightly clearance of fibrillar αSN by human monocytes. The induced change coupled concomitantly with phagolysosome formation. Data mining of the brain transcriptome in Parkinson disease patients supported CR4 as an active αSN clearance mechanism in this disease. Our results associate an important part of the innate immune system, namely complement receptors, with the central molecular mechanisms of CNS protein aggregation in neurodegenerative disorders.


Asunto(s)
Integrina alfaXbeta2 , Macrófagos , Enfermedad de Parkinson , Fagosomas , Agregación Patológica de Proteínas , alfa-Sinucleína , Humanos , Integrina alfaXbeta2/química , Integrina alfaXbeta2/genética , Integrina alfaXbeta2/inmunología , Macrófagos/inmunología , Macrófagos/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología , Fagosomas/química , Fagosomas/genética , Fagosomas/inmunología , Fagosomas/patología , Agregación Patológica de Proteínas/genética , Agregación Patológica de Proteínas/inmunología , Agregación Patológica de Proteínas/patología , Estructura Cuaternaria de Proteína , alfa-Sinucleína/química , alfa-Sinucleína/genética , alfa-Sinucleína/inmunología
19.
Stem Cell Res ; 41: 101645, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31759291

RESUMEN

Porcine embryonic and induced pluripotent stem cells (ESCs; iPSCs) have proven difficult to derive and maintain in vitro. This may be due to inappropriate culturing conditions and incomplete activation of proper pluripotency networks. To this end, we characterized the transcriptome of porcine inner cell mass, epiblast, and transgene-dependent iPSCs in relation to human and mouse embryonic and epiblast stem cells. We found that porcine inner cell mass has a unique pluripotency transcriptome distinct from human and mouse ESCs but shares more features with human naïve-like than primed stem cell states, as illustrated by their expression of KLF17 but not KLF2. Our data further show that current reprogramming strategies fail to silence parental fibroblast-specific genes and to activate specific signalling pathways that may be important for porcine pluripotency. Accordingly, we used human naïve culturing conditions to improve reprogramming efficiencies of porcine embryonic fibroblasts and enable essential naïve stem cell markers such as NANOG, KLF17 and CDH1to be expressed. The resultant porcine iPSC-like cells display a transcriptomic signature more closely resembling an inner cell mass state. These results represent new important steps towards generating bona fide porcine iPSCs and their great potential in translational medicine.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Diferenciación Celular , Perfilación de la Expresión Génica , Células Madre Pluripotentes Inducidas/metabolismo , Transcriptoma , Animales , Masa Celular Interna del Blastocisto/citología , Masa Celular Interna del Blastocisto/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Especificidad de la Especie , Porcinos
20.
Nat Neurosci ; 22(7): 1066-1074, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209380

RESUMEN

Cannabis is the most frequently used illicit psychoactive substance worldwide; around one in ten users become dependent. The risk for cannabis use disorder (CUD) has a strong genetic component, with twin heritability estimates ranging from 51 to 70%. Here we performed a genome-wide association study of CUD in 2,387 cases and 48,985 controls, followed by replication in 5,501 cases and 301,041 controls. We report a genome-wide significant risk locus for CUD (P = 9.31 × 10-12) that replicates in an independent population (Preplication = 3.27 × 10-3, Pmeta-analysis = 9.09 × 10-12). The index variant (rs56372821) is a strong expression quantitative trait locus for cholinergic receptor nicotinic α2 subunit (CHRNA2); analyses of the genetically regulated gene expression identified a significant association of CHRNA2 expression with CUD in brain tissue. At the polygenic level, analyses revealed a significant decrease in the risk of CUD with increased load of variants associated with cognitive performance. The results provide biological insights and inform on the genetic architecture of CUD.


Asunto(s)
Abuso de Marihuana/genética , Proteínas del Tejido Nervioso/fisiología , Receptores Nicotínicos/fisiología , Edad de Inicio , Alelos , Trastorno por Déficit de Atención con Hiperactividad/genética , Encéfalo/metabolismo , Estudios de Casos y Controles , Cromosomas Humanos Par 8/genética , Cognición/fisiología , Estudios de Cohortes , Factores de Confusión Epidemiológicos , Dinamarca , Escolaridad , Femenino , Perfilación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Islandia , Masculino , Herencia Multifactorial , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Receptores Nicotínicos/biosíntesis , Receptores Nicotínicos/genética , Esquizofrenia/genética , Fumar/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...