Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 15(11): 2315-22, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27268504

ABSTRACT

The recent Zika virus (ZIKV) outbreak in the Western hemisphere is associated with severe pathology in newborns, including microcephaly and brain damage. The mechanisms underlying these outcomes are under intense investigation. Here, we show that a 2015 ZIKV isolate replicates in multiple cell types, including primary human fetal neural progenitors (hNPs). In immortalized cells, ZIKV is cytopathic and grossly rearranges endoplasmic reticulum membranes similar to other flaviviruses. In hNPs, ZIKV infection has a partial cytopathic phase characterized by cell rounding, pyknosis, and activation of caspase 3. Despite notable cell death, ZIKV did not activate a cytokine response in hNPs. This lack of cell intrinsic immunity to ZIKV is consistent with our observation that virus replication persists in hNPs for at least 28 days. These findings, supported by published fetal neuropathology, establish a proof-of-concept that neural progenitors in the developing human fetus can be direct targets of detrimental ZIKV-induced pathology.


Subject(s)
Cytopathogenic Effect, Viral/immunology , Fetus/pathology , Neural Stem Cells/immunology , Neural Stem Cells/virology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Cell Line , Humans , Time Factors , Virus Replication , Zika Virus/isolation & purification , Zika Virus/physiology , Zika Virus/ultrastructure
2.
Neuron ; 83(1): 69-86, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-24991955

ABSTRACT

Neural stem cells have been adopted to model a wide range of neuropsychiatric conditions in vitro. However, how well such models correspond to in vivo brain has not been evaluated in an unbiased, comprehensive manner. We used transcriptomic analyses to compare in vitro systems to developing human fetal brain and observed strong conservation of in vivo gene expression and network architecture in differentiating primary human neural progenitor cells (phNPCs). Conserved modules are enriched in genes associated with ASD, supporting the utility of phNPCs for studying neuropsychiatric disease. We also developed and validated a machine learning approach called CoNTExT that identifies the developmental maturity and regional identity of in vitro models. We observed strong differences between in vitro models, including hiPSC-derived neural progenitors from multiple laboratories. This work provides a systems biology framework for evaluating in vitro systems and supports their value in studying the molecular mechanisms of human neurodevelopmental disease.


Subject(s)
Artificial Intelligence , Cerebral Cortex/embryology , Embryonic Stem Cells/physiology , Gene Regulatory Networks/genetics , Models, Neurological , Neural Stem Cells/physiology , Artificial Intelligence/trends , Cells, Cultured , Cerebral Cortex/cytology , Female , Humans , Male
3.
Neuron ; 72(4): 501-3, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-22099453

ABSTRACT

Two papers address the contribution of DISC1 to neural development and schizophrenia risk in this issue of Neuron. These complementary studies elegantly bridge the gap between genetic and cellular studies of schizophrenia, providing a level of functional validation that is often lacking in the field.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Brain Chemistry/genetics , Brain/growth & development , Glycogen Synthase Kinase 3/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Polymorphism, Genetic/genetics , Schizophrenia/metabolism , Signal Transduction/genetics , Wnt3A Protein/genetics , Animals , Female , Glycogen Synthase Kinase 3 beta , Humans , Male , Pregnancy
4.
Sci Signal ; 4(193): ra65, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21971039

ABSTRACT

Wnt proteins are critical to mammalian brain development and function. The canonical Wnt signaling pathway involves the stabilization and nuclear translocation of ß-catenin; however, Wnt also signals through alternative, noncanonical pathways. To gain a systems-level, genome-wide view of Wnt signaling, we analyzed Wnt1-stimulated changes in gene expression by transcriptional microarray analysis in cultured human neural progenitor (hNP) cells at multiple time points over a 72-hour time course. We observed a widespread oscillatory-like pattern of changes in gene expression, involving components of both the canonical and the noncanonical Wnt signaling pathways. A higher-order, systems-level analysis that combined independent component analysis, waveform analysis, and mutual information-based network construction revealed effects on pathways related to cell death and neurodegenerative disease. Wnt effectors were tightly clustered with presenilin1 (PSEN1) and granulin (GRN), which cause dominantly inherited forms of Alzheimer's disease and frontotemporal dementia (FTD), respectively. We further explored a potential link between Wnt1 and GRN and found that Wnt1 decreased GRN expression by hNPs. Conversely, GRN knockdown increased WNT1 expression, demonstrating that Wnt and GRN reciprocally regulate each other. Finally, we provided in vivo validation of the in vitro findings by analyzing gene expression data from individuals with FTD. These unbiased and genome-wide analyses provide evidence for a connection between Wnt signaling and the transcriptional regulation of neurodegenerative disease genes.


Subject(s)
Alzheimer Disease/metabolism , Frontotemporal Dementia/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/biosynthesis , Transcription, Genetic , Wnt Signaling Pathway , Wnt1 Protein/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Cells, Cultured , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Gene Expression Profiling , Genome-Wide Association Study , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Presenilin-1/biosynthesis , Presenilin-1/genetics , Progranulins , Wnt1 Protein/genetics
5.
Neuron ; 71(6): 1030-42, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21943601

ABSTRACT

Progranulin (GRN) mutations cause frontotemporal dementia (FTD), but GRN's function in the CNS remains largely unknown. To identify the pathways downstream of GRN, we used weighted gene coexpression network analysis (WGCNA) to develop a systems-level view of transcriptional alterations in a human neural progenitor model of GRN-deficiency. This highlighted key pathways such as apoptosis and ubiquitination in GRN deficient human neurons, while revealing an unexpected major role for the Wnt signaling pathway, which was confirmed by analysis of gene expression data from postmortem FTD brain. Furthermore, we observed that the Wnt receptor Fzd2 was one of only a few genes upregulated at 6 weeks in a GRN knockout mouse, and that FZD2 reduction caused increased apoptosis, while its upregulation promoted neuronal survival in vitro. Together, these in vitro and in vivo data point to an adaptive role for altered Wnt signaling in GRN deficiency-mediated FTD, representing a potential therapeutic target.


Subject(s)
Genome , Genomics/methods , Intercellular Signaling Peptides and Proteins/deficiency , Signal Transduction/physiology , Wnt Proteins/metabolism , Animals , Cell Death , Cell Differentiation , Cells, Cultured , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Microarray Analysis , Neural Stem Cells/physiology , Neurons/physiology , Progranulins , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/genetics
6.
Stem Cells ; 27(5): 1130-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19418460

ABSTRACT

Wnt signaling regulates neural stem cell (NSC) function and development throughout an individual's lifetime. Intriguingly, adult hippocampal progenitors (AHPs) produce several Wnts, and the intracellular machinery necessary to respond to them, creating the potential for an active autocrine-signaling loop within this stem cell niche. However, the standard luciferase-based Wnt assay failed to detect this signaling loop. This assay is inherently less temporally sensitive to activity among a population of unsynchronized proliferating cells because it relies on the rapidly degrading reporter luciferase. We circumvented this limitation using a promoter assay that employs green fluorescent protein (GFP), as a relatively long-lived reporter of canonical Wnt activity. We found that at baseline, AHPs secreted functional Wnt that self-stimulates low-level canonical Wnt signaling. Elimination baseline Wnt activity, via application of an extracellular Wnt antagonist promoted neurogenesis, based on a combination of unbiased gene expression analysis and cell-fate analysis. A detailed clonal analysis of progenitors transduced with specific intracellular antagonists of canonical signaling, either Axin or truncated cadherin (beta-catenin sequestering), revealed that loss of baseline signaling depletes the population of multipotent precursors, thereby driving an increasing fraction to assume a committed cell fate (i.e., unipotent progenitors). Similarly, baseline Wnt signaling repressed differentiation of human NSCs. Although the specific Wnts produced by neural precursors vary with age and between species, their effects remain remarkably consistent. In sum, this study establishes that autonomous Wnt signaling is a conserved feature of the neurogenic niche that preserves the delicate balance between NSC maintenance and differentiation.


Subject(s)
Neurons/cytology , Neurons/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Animals , Autocrine Communication , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Hippocampus/cytology , Humans , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Rats , Species Specificity , beta Catenin/metabolism
8.
Neurobiol Dis ; 22(2): 302-11, 2006 May.
Article in English | MEDLINE | ID: mdl-16442805

ABSTRACT

Friedreich's ataxia (FRDA) is caused by reduction of frataxin levels to 5-35%. To better understand the biochemical sequelae of frataxin reduction, in absence of the confounding effects of neurodegeneration, we studied the gene expression profile of a mouse model expressing 25-36% of the normal frataxin levels, and not showing a detectable phenotype or neurodegenerative features. Despite having no overt phenotype, a clear microarray gene expression phenotype was observed. This phenotype followed the known regional susceptibility in this disease, most changes occurring in the spinal cord. Additionally, gene ontology analysis identified a clear mitochondrial component, consistent with previous findings. We were able to confirm a subset of changes in fibroblast cell lines from patients. The identification of a core set of genes changing early in the FRDA pathogenesis can be a useful tool in both clarifying the disease process and in evaluating new therapeutic strategies.


Subject(s)
Central Nervous System/metabolism , Friedreich Ataxia/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Iron-Binding Proteins/genetics , Nerve Degeneration/genetics , Animals , Cell Line , Central Nervous System/pathology , Central Nervous System/physiopathology , Disease Models, Animal , Female , Friedreich Ataxia/metabolism , Friedreich Ataxia/physiopathology , Gene Expression Profiling , Humans , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Oligonucleotide Array Sequence Analysis , Phenotype , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL
...