Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38496540

RESUMEN

Glioblastoma (GBM), a universally fatal brain cancer, infiltrates the brain and can be synaptically innervated by neurons, which drives tumor progression 1-6 . Synaptic inputs onto GBM cells identified so far are largely short-range and glutamatergic 7-9 . The extent of integration of GBM cells into brain-wide neuronal circuitry is not well understood. Here we applied a rabies virus-mediated retrograde monosynaptic tracing approach 10-12 to systematically investigate circuit integration of human GBM organoids transplanted into adult mice. We found that GBM cells from multiple patients rapidly integrated into brain-wide neuronal circuits and exhibited diverse local and long-range connectivity. Beyond glutamatergic inputs, we identified a variety of neuromodulatory inputs across the brain, including cholinergic inputs from the basal forebrain. Acute acetylcholine stimulation induced sustained calcium oscillations and long-lasting transcriptional reprogramming of GBM cells into a more invasive state via the metabotropic CHRM3 receptor. CHRM3 downregulation suppressed GBM cell invasion, proliferation, and survival in vitro and in vivo. Together, these results reveal the capacity of human GBM cells to rapidly and robustly integrate into anatomically and molecularly diverse neuronal circuitry in the adult brain and support a model wherein rapid synapse formation onto GBM cells and transient activation of upstream neurons may lead to a long-lasting increase in fitness to promote tumor infiltration and progression.

2.
Cell Rep ; 42(4): 112334, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37043350

RESUMEN

Hippocampal place cells exhibit spatially modulated firing, or place fields, which can remap to encode changes in the environment or other variables. Unique among hippocampal subregions, the dentate gyrus (DG) has two excitatory populations of place cells, granule cells and mossy cells, which are among the least and most active spatially modulated cells in the hippocampus, respectively. Previous studies of remapping in the DG have drawn different conclusions about whether granule cells exhibit global remapping and contribute to the encoding of context specificity. By recording granule cells and mossy cells as mice foraged in different environments, we found that by most measures, both granule cells and mossy cells remapped robustly but through different mechanisms that are consistent with firing properties of each cell type. Our results resolve the ambiguity surrounding remapping in the DG and suggest that most spatially modulated granule cells contribute to orthogonal representations of distinct spatial contexts.


Asunto(s)
Fibras Musgosas del Hipocampo , Células de Lugar , Ratones , Animales , Giro Dentado/metabolismo , Hipocampo
3.
Curr Opin Neurobiol ; 79: 102672, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36634408

RESUMEN

Human pluripotent stem cells can be differentiated into cell types that are representative of the central nervous system. Under specific culture conditions, these cells can be induced to self-organize into 3D organoids that are reminiscent of the developing brain. Microglia are the resident immune cells of the brain but are derived from a different lineage than neural cells, which presents a challenge to modeling neuroimmune interactions. Although human microglia-like cells can be differentiated from pluripotent stem cells, important considerations include ensuring the identity of microglia, which can be influenced by both the lineage and the local environment, and developing culture methods that promote the integration and survival of diverse cell types in a physiologically relevant model. Recently, several strategies to generate neural organoids with integrated microglia have been demonstrated and provide new opportunities to interrogate interactions among microglia and neurons during development and in response to injury and disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Neuroinmunomodulación , Células Madre Pluripotentes Inducidas/metabolismo , Encéfalo , Sistema Nervioso Central , Diferenciación Celular
4.
Front Virol ; 22022.
Artículo en Inglés | MEDLINE | ID: mdl-36325520

RESUMEN

Understanding the impact of viral pathogens on the human central nervous system (CNS) has been challenging due to the lack of viable human CNS models for controlled experiments to determine the causal factors underlying pathogenesis. Human embryonic stem cells (ESCs) and, more recently, cellular reprogramming of adult somatic cells to generate human induced pluripotent stem cells (iPSCs) provide opportunities for directed differentiation to neural cells that can be used to evaluate the impact of known and emerging viruses on neural cell types. Pluripotent stem cells (PSCs) can be induced to neural lineages in either two- (2D) or three-dimensional (3D) cultures, each bearing distinct advantages and limitations for modeling viral pathogenesis and evaluating effective therapeutics. Here we review the current state of technology in stem cell-based modeling of the CNS and how these models can be used to determine viral tropism and identify cellular phenotypes to investigate virus-host interactions and facilitate drug screening. We focus on several viruses (e.g., human immunodeficiency virus (HIV), herpes simplex virus (HSV), Zika virus (ZIKV), human cytomegalovirus (HCMV), SARS-CoV-2, West Nile virus (WNV)) to illustrate key advantages, as well as challenges, of PSC-based models. We also discuss how human PSC-based models can be used to evaluate the safety and efficacy of therapeutic drugs by generating data that are complementary to existing preclinical models. Ultimately, these efforts could facilitate the movement towards personalized medicine and provide patients and physicians with an additional source of information to consider when evaluating available treatment strategies.

5.
Curr Biol ; 32(5): 1088-1101.e5, 2022 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-35108522

RESUMEN

The hippocampus is involved in the formation of memories that require associations among stimuli to construct representations of space and the items and events within that space. Neurons in the dentate gyrus (DG), an initial input region of the hippocampus, have robust spatial tuning, but it is unclear how nonspatial information may be integrated with spatial activity in this region. We recorded from the DG of 21 adult mice as they foraged for food in an environment that contained discrete objects. We found DG cells with multiple firing fields at a fixed distance and direction from objects (landmark vector cells) and cells that exhibited localized changes in spatial firing when objects in the environment were manipulated. By classifying recorded DG cells into putative dentate granule cells and mossy cells, we examined how the addition or displacement of objects affected the spatial firing of these DG cell types. Object-related activity was detected in a significant proportion of mossy cells. Although few granule cells with responses to object manipulations were recorded, likely because of the sparse nature of granule cell firing, there was generally no significant difference in the proportion of granule cells and mossy cells with object responses. When mice explored a second environment with the same objects, DG spatial maps completely reorganized, and a different subset of cells responded to object manipulations. Together, these data reveal the capacity of DG cells to detect small changes in the environment while preserving a stable spatial representation of the overall context.


Asunto(s)
Hipocampo , Neuronas , Animales , Giro Dentado/fisiología , Hipocampo/fisiología , Ratones , Neuronas/fisiología
6.
J Mol Biol ; 434(3): 167243, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34536442

RESUMEN

Brain organoids are self-organized three-dimensional aggregates generated from pluripotent stem cells. They exhibit complex cell diversities and organized architectures that resemble human brain development ranging from neural tube formation, neuroepithelium differentiation, neurogenesis and gliogenesis, to neural circuit formation. Rapid advancements in brain organoid culture technologies have allowed researchers to generate more accurate models of human brain development and neurological diseases. These models also allow for direct investigation of pathological processes associated with infectious diseases affecting the nervous system. In this review, we first briefly summarize recent advancements in brain organoid methodologies and neurodevelopmental processes that can be effectively modeled by brain organoids. We then focus on applications of brain organoids to investigate the pathogenesis of neurotropic viral infection. Finally, we discuss limitations of the current brain organoid methodologies as well as applications of other organ specific organoids in the infectious disease research.


Asunto(s)
Encéfalo , Enfermedades Virales del Sistema Nervioso Central , Organoides , Encéfalo/crecimiento & desarrollo , Encéfalo/virología , Enfermedades Virales del Sistema Nervioso Central/virología , Humanos , Neurogénesis , Organoides/virología
8.
Biol Psychiatry ; 92(10): 815-826, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34247782

RESUMEN

BACKGROUND: Gene dosage imbalance caused by copy number variations (CNVs) is a prominent contributor to brain disorders. In particular, 15q11.2 CNV duplications and deletions have been associated with autism spectrum disorder and schizophrenia, respectively. The mechanism underlying these diametric contributions remains unclear. METHODS: We established both loss-of-function and gain-of-function mouse models of Cyfip1, one of four genes within 15q11.2 CNVs. To assess the functional consequences of altered CYFIP1 levels, we performed systematic investigations on behavioral, electrophysiological, and biochemical phenotypes in both mouse models. In addition, we utilized RNA immunoprecipitation sequencing (RIP-seq) analysis to reveal molecular targets of CYFIP1 in vivo. RESULTS: Cyfip1 loss-of-function and gain-of function mouse models exhibited distinct and shared behavioral abnormalities related to autism spectrum disorder and schizophrenia. RIP-seq analysis identified messenger RNA targets of CYFIP1 in vivo, including postsynaptic NMDA receptor (NMDAR) complex components. In addition, these mouse models showed diametric changes in levels of postsynaptic NMDAR complex components at synapses because of dysregulated protein translation, resulting in bidirectional alteration of NMDAR-mediated signaling. Importantly, pharmacological balancing of NMDAR signaling in these mouse models with diametric Cyfip1 dosages rescues behavioral abnormalities. CONCLUSIONS: CYFIP1 regulates protein translation of NMDAR and associated complex components at synapses to maintain normal synaptic functions and behaviors. Our integrated analyses provide insight into how gene dosage imbalance caused by CNVs may contribute to divergent neuropsychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastornos Mentales , Ratones , Animales , Receptores de N-Metil-D-Aspartato/genética , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Variaciones en el Número de Copia de ADN , Ratones Endogámicos C57BL , N-Metilaspartato/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Modelos Animales de Enfermedad , ARN Mensajero , ARN
10.
Nat Commun ; 12(1): 1398, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33658519

RESUMEN

We previously identified a causal link between a rare patient mutation in DISC1 (disrupted-in-schizophrenia 1) and synaptic deficits in cortical neurons differentiated from isogenic patient-derived induced pluripotent stem cells (iPSCs). Here we find that transcripts related to phosphodiesterase 4 (PDE4) signaling are significantly elevated in human cortical neurons differentiated from iPSCs with the DISC1 mutation and that inhibition of PDE4 or activation of the cAMP signaling pathway functionally rescues synaptic deficits. We further generated a knock-in mouse line harboring the same patient mutation in the Disc1 gene. Heterozygous Disc1 mutant mice exhibit elevated levels of PDE4s and synaptic abnormalities in the brain, and social and cognitive behavioral deficits. Pharmacological inhibition of the PDE4 signaling pathway rescues these synaptic, social and cognitive behavioral abnormalities. Our study shows that patient-derived isogenic iPSC and humanized mouse disease models are integral and complementary for translational studies with a better understanding of underlying molecular mechanisms.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Inhibidores de Fosfodiesterasa 4/farmacología , Esquizofrenia/genética , Animales , Conducta Animal/efectos de los fármacos , Corteza Cerebral/fisiología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Humanos , Masculino , Ratones Mutantes , Mutación , Neuronas/efectos de los fármacos , Rolipram/farmacología , Esquizofrenia/patología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología
11.
Genomics Proteomics Bioinformatics ; 19(1): 108-122, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33610792

RESUMEN

The Zika virus (ZIKV) and dengue virus (DENV) flaviviruses exhibit similar replicative processes but have distinct clinical outcomes. A systematic understanding of virus-host protein-protein interaction networks can reveal cellular pathways critical to viral replication and disease pathogenesis. Here we employed three independent systems biology approaches toward this goal. First, protein array analysis of direct interactions between individual ZIKV/DENV viral proteins and 20,240 human proteins revealed multiple conserved cellular pathways and protein complexes, including proteasome complexes. Second, an RNAi screen of 10,415 druggable genes identified the host proteins required for ZIKV infection and uncovered that proteasome proteins were crucial in this process. Third, high-throughput screening of 6016 bioactive compounds for ZIKV inhibition yielded 134 effective compounds, including six proteasome inhibitors that suppress both ZIKV and DENV replication. Integrative analyses of these orthogonal datasets pinpoint proteasomes as critical host machinery for ZIKV/DENV replication. Our study provides multi-omics datasets for further studies of flavivirus-host interactions, disease pathogenesis, and new drug targets.


Asunto(s)
Virus del Dengue , Dengue/genética , Complejo de la Endopetidasa Proteasomal , Infección por el Virus Zika , Virus Zika , Virus del Dengue/genética , Virus del Dengue/fisiología , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Biología de Sistemas , Replicación Viral , Virus Zika/genética , Virus Zika/fisiología , Infección por el Virus Zika/genética
12.
Mol Psychiatry ; 26(4): 1346-1360, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-31444471

RESUMEN

Psychiatric disorders are a collection of heterogeneous mental disorders arising from a contribution of genetic and environmental insults, many of which molecularly converge on transcriptional dysregulation, resulting in altered synaptic functions. The underlying mechanisms linking the genetic lesion and functional phenotypes remain largely unknown. Patient iPSC-derived neurons with a rare frameshift DISC1 (Disrupted-in-schizophrenia 1) mutation have previously been shown to exhibit aberrant gene expression and deficits in synaptic functions. How DISC1 regulates gene expression is largely unknown. Here we show that Activating Transcription Factor 4 (ATF4), a DISC1 binding partner, is more abundant in the nucleus of DISC1 mutant human neurons and exhibits enhanced binding to a collection of dysregulated genes. Functionally, overexpressing ATF4 in control neurons recapitulates deficits seen in DISC1 mutant neurons, whereas transcriptional and synaptic deficits are rescued in DISC1 mutant neurons with CRISPR-mediated heterozygous ATF4 knockout. By solving the high-resolution atomic structure of the DISC1-ATF4 complex, we show that mechanistically, the mutation of DISC1 disrupts normal DISC1-ATF4 interaction, and results in excessive ATF4 binding to DNA targets and deregulated gene expression. Together, our study identifies the molecular and structural basis of an DISC1-ATF4 interaction underlying transcriptional and synaptic dysregulation in an iPSC model of mental disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas , Trastornos Mentales , Factor de Transcripción Activador 4/genética , Humanos , Proteínas del Tejido Nervioso/genética , Neuronas
13.
J Neuroimmune Pharmacol ; 16(1): 113-129, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-31512167

RESUMEN

As antiretroviral therapy (ART) becomes increasingly affordable and accessible to women of childbearing age across the globe, the number of children who are exposed to Human Immunodeficiency Viruses (HIV) but remain uninfected is on the rise, almost all of whom were also exposed to ART perinatally. Although ART has successfully aided in the decline of mother-to-child-transmission of HIV, the long-term effects of in utero exposure to ART on fetal and postnatal neurodevelopment remain unclear. Evaluating the safety and efficacy of therapeutic drugs for pregnant women is a challenge due to the historic limitations on their inclusion in clinical trials and the dynamic physiological states during pregnancy that can alter the pharmacokinetics of drug metabolism and fetal drug exposure. Thus, much of our data on the potential consequences of ART drugs on the developing nervous system comes from preclinical animal models and clinical observational studies. In this review, we will discuss the current state of knowledge and existing approaches to investigate whether ART affects fetal brain development, and describe novel human stem cell-based strategies that may provide additional information to better predict the impact of specific drugs on the human central nervous system. Graphical Abstract Approaches to evaluate the impact of drugs on the developing brain. Dysregulation of the developing nervous system can lead to long-lasting changes. Integration of data from animal models, clinical observations, and cell culture studies is needed to predict the safety of therapeutic antiretroviral drugs during pregnancy. New approaches include human induced pluripotent stem cell (iPSC)-based 2D and 3D models of neuronal networks and brain regions, as well as single cell profiling in response to drug exposure.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Trastornos del Neurodesarrollo/inducido químicamente , Adulto , Animales , Fármacos Anti-VIH/farmacocinética , Fármacos Anti-VIH/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Técnicas de Cultivo de Célula , División Celular , Femenino , Feto/efectos de los fármacos , Predicción , Infecciones por VIH/congénito , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/transmisión , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Intercambio Materno-Fetal , Ratones , Mitocondrias/efectos de los fármacos , Modelos Animales , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Estrés Oxidativo , Guías de Práctica Clínica como Asunto , Embarazo , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Efectos Tardíos de la Exposición Prenatal , Sinapsis
14.
Adv Neurobiol ; 25: 237-257, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32578150

RESUMEN

Psychiatric disorders are among the most challenging human diseases to understand at a mechanistic level due to the heterogeneity of symptoms within established diagnostic categories, the general absence of focal pathology, and the genetic complexity inherent in these mostly polygenic disorders. Each of these features presents unique challenges to disease modeling for biological discovery, drug development, or improved diagnostics. In addition, live human neural tissue has been largely inaccessible to experimentation, leaving gaps in our knowledge derived from animal models that cannot fully recapitulate the features of the disease, indirect measures of brain function in human patients, and from analyses of postmortem tissue that can be confounded by comorbid conditions and medication history.


Asunto(s)
Células Madre Pluripotentes Inducidas , Trastornos Mentales , Animales , Humanos
15.
Behav Brain Res ; 379: 112346, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31722241

RESUMEN

Hypotheses about the functional properties of the dentate gyrus and adult dentate neurogenesis have been shaped by early observations of the anatomy of this region, mostly in rodents. This has led to the development of a few core propositions that have guided research over the past several years, including the predicted role of this region in pattern separation and the local transformation of inputs from the entorhinal cortex. We now have the opportunity to review these predictions and update these anatomical observations based on recently developed techniques that reveal the complex structure, connectivity, and dynamic properties of distinct cell populations in the dentate gyrus at a higher resolution. Cumulative evidence suggests that the dentate gyrus and adult-born granule cells play a role in some forms of behavioral discriminations, but there are still many unanswered questions about how the dentate gyrus processes information to support the disambiguation of stimuli.


Asunto(s)
Giro Dentado/anatomía & histología , Giro Dentado/fisiología , Discriminación en Psicología/fisiología , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Animales
16.
Cell ; 180(1): 188-204.e22, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31883794

RESUMEN

Glioblastomas exhibit vast inter- and intra-tumoral heterogeneity, complicating the development of effective therapeutic strategies. Current in vitro models are limited in preserving the cellular and mutational diversity of parental tumors and require a prolonged generation time. Here, we report methods for generating and biobanking patient-derived glioblastoma organoids (GBOs) that recapitulate the histological features, cellular diversity, gene expression, and mutational profiles of their corresponding parental tumors. GBOs can be generated quickly with high reliability and exhibit rapid, aggressive infiltration when transplanted into adult rodent brains. We further demonstrate the utility of GBOs to test personalized therapies by correlating GBO mutational profiles with responses to specific drugs and by modeling chimeric antigen receptor T cell immunotherapy. Our studies show that GBOs maintain many key features of glioblastomas and can be rapidly deployed to investigate patient-specific treatment strategies. Additionally, our live biobank establishes a rich resource for basic and translational glioblastoma research.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glioblastoma/metabolismo , Organoides/crecimiento & desarrollo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Bancos de Muestras Biológicas , Femenino , Glioblastoma/genética , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Modelos Biológicos , Organoides/metabolismo , Reproducibilidad de los Resultados , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
17.
Cell Rep ; 28(6): 1419-1428.e3, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31390557

RESUMEN

Excitation-inhibition (E-I) imbalance is considered a hallmark of various neurodevelopmental disorders, including schizophrenia and autism. How genetic risk factors disrupt coordinated glutamatergic and GABAergic synapse formation to cause an E-I imbalance is not well understood. Here, we show that knockdown of Disrupted-in-schizophrenia 1 (DISC1), a risk gene for major mental disorders, leads to E-I imbalance in mature dentate granule neurons. We found that excessive GABAergic inputs from parvalbumin-, but not somatostatin-, expressing interneurons enhance the formation of both glutamatergic and GABAergic synapses in immature mutant neurons. Following the switch in GABAergic signaling polarity from depolarizing to hyperpolarizing during neuronal maturation, heightened inhibition from excessive parvalbumin+ GABAergic inputs causes loss of excitatory glutamatergic synapses in mature mutant neurons, resulting in an E-I imbalance. Our findings provide insights into the developmental role of depolarizing GABA in establishing E-I balance and how it can be influenced by genetic risk factors for mental disorders.


Asunto(s)
Predisposición Genética a la Enfermedad , Trastornos Mentales/genética , Neuronas/fisiología , Sinapsis/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Polaridad Celular , Femenino , Neuronas GABAérgicas/fisiología , Técnicas de Silenciamiento del Gen , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/fisiología , Inhibición Neural , Neurogénesis/genética , Neurogénesis/fisiología , Factores de Riesgo , Sinapsis/genética , Potenciales Sinápticos
18.
Annu Rev Neurosci ; 42: 249-269, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31283901

RESUMEN

In 2015, public awareness of Zika virus (ZIKV) rose in response to alarming statistics of infants with microcephaly being born to women who were infected with the virus during pregnancy, triggering global concern over these potentially devastating consequences. Although we have discovered a great deal about the genome and pathogenesis of this reemergent flavivirus since this recent outbreak, we still have much more to learn, including the nature of the virus-host interactions and mechanisms that determine its tropism and pathogenicity in the nervous system, which are in turn shaped by the continual evolution of the virus. Inevitably, we will find out more about the potential long-term effects of ZIKV exposure on the nervous system from ongoing longitudinal studies. Integrating clinical and epidemiological data with a wider range of animal and human cell culture models will be critical to understanding the pathogenetic mechanisms and developing more specific antiviral compounds and vaccines.


Asunto(s)
Enfermedades del Sistema Nervioso/virología , Infección por el Virus Zika/fisiopatología , Adulto , Animales , Encéfalo/embriología , Encéfalo/patología , Encéfalo/virología , Células Cultivadas , Enfermedades Transmisibles Emergentes , Brotes de Enfermedades , Femenino , Regulación del Desarrollo de la Expresión Génica , Regulación Viral de la Expresión Génica , Vectores Genéticos/genética , Interacciones Microbiota-Huesped , Humanos , Recién Nacido , Macaca mulatta , Ratones , Microbiota , Microcefalia/embriología , Microcefalia/etiología , Microcefalia/virología , Microglía/fisiología , Modelos Animales , Enfermedades del Sistema Nervioso/fisiopatología , Neurogénesis , Embarazo , Complicaciones Infecciosas del Embarazo/fisiopatología , Receptores Virales/fisiología , Estudios en Gemelos como Asunto , Vacunas Virales , Virus Zika/inmunología , Virus Zika/aislamiento & purificación , Virus Zika/patogenicidad , Virus Zika/fisiología , Infección por el Virus Zika/diagnóstico , Infección por el Virus Zika/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA