Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (206)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38682940

ABSTRACT

Resolutive cures for spinal cord injuries (SCIs) are still lacking, due to the complex pathophysiology. One of the most promising regenerative approaches is based on stem cell transplantation to replace lost tissue and promote functional recovery. This approach should be further explored better in vitro and ex vivo for safety and efficacy before proceeding with more expensive and time-consuming animal testing. In this work, we show the establishment of a long-term platform based on mouse spinal cord (SC) organotypic slices transplanted with human neural stem cells to test cellular replacement therapies for SCIs. Standard SC organotypic cultures are maintained for around 2 or 3 weeks in vitro. Here, we describe an optimized protocol for long-term maintenance (≥30 days) for up to 90 days. The medium used for long-term culturing of SC slices was also optimized for transplanting neural stem cells into the organotypic model. Human SC-derived neuroepithelial stem (h-SC-NES) cells carrying a green fluorescent protein (GFP) reporter were transplanted into mouse SC slices. Thirty days after the transplant, cells still show GFP expression and a low apoptotic rate, suggesting that the optimized environment sustained their survival and integration inside the tissue. This protocol represents a robust reference for efficiently testing cell replacement therapies in the SC tissue. This platform will allow researchers to perform an ex vivo pre-screening of different cell transplantation therapies, helping them to choose the most appropriate strategy before proceeding with in vivo experiments.


Subject(s)
Neural Stem Cells , Spinal Cord Injuries , Spinal Cord , Animals , Mice , Spinal Cord Injuries/therapy , Humans , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Spinal Cord/cytology , Organ Culture Techniques/methods , Stem Cell Transplantation/methods
2.
Antiviral Res ; 223: 105816, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38286212

ABSTRACT

Human cytomegalovirus (HCMV) is the viral leading cause of congenital defects in newborns worldwide. Many aspects of congenital CMV (cCMV) infection, which currently lacks a specific treatment, as well as the main determinants of neuropathogenesis in the developing brain during HCMV infection are unclear. In this study, we modeled HCMV infection at different stages of neural development. Moreover, we evaluated the effects of both approved and investigational anti-HCMV drugs on viral replication and gene expression in two different neural progenitor cell lines, i.e., human embryonic stem cells-derived neural stem cells (NSCs) and fetus-derived neuroepithelial stem (NES) cells. Ganciclovir, letermovir, nitazoxanide, and the ozonide OZ418 reduced viral DNA synthesis and the production of infectious virus in both lines of neural progenitors. HCMV infection dysregulated the expression of genes that either are markers of neural progenitors, such as SOX2, NESTIN, PAX-6, or play a role in neurogenesis, such as Doublecortin. Treatment with antiviral drugs had different effects on HCMV-induced dysregulation of the genes under investigation. This study contributes to the understanding of the molecular mechanisms of cCMV neuropathogenesis and paves the way for further consideration of anti-HCMV drugs as candidate therapeutic agents for the amelioration of cCMV-associated neurological manifestations.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Infant, Newborn , Humans , Cytomegalovirus Infections/drug therapy , Brain , Drugs, Investigational , Stem Cells , Antiviral Agents/pharmacology
4.
Antiviral Res ; 216: 105664, 2023 08.
Article in English | MEDLINE | ID: mdl-37414288

ABSTRACT

Recent evidence suggests that lipids play a crucial role in viral infections beyond their traditional functions of supplying envelope and energy, and creating protected niches for viral replication. In the case of Zika virus (ZIKV), it alters host lipids by enhancing lipogenesis and suppressing ß-oxidation to generate viral factories at the endoplasmic reticulum (ER) interface. This discovery prompted us to hypothesize that interference with lipogenesis could serve as a dual antiviral and anti-inflammatory strategy to combat the replication of positive sense single-stranded RNA (ssRNA+) viruses. To test this hypothesis, we examined the impact of inhibiting N-Acylethanolamine acid amidase (NAAA) on ZIKV-infected human Neural Stem Cells. NAAA is responsible for the hydrolysis of palmitoylethanolamide (PEA) in lysosomes and endolysosomes. Inhibition of NAAA results in PEA accumulation, which activates peroxisome proliferator-activated receptor-α (PPAR-α), directing ß-oxidation and preventing inflammation. Our findings indicate that inhibiting NAAA through gene-editing or drugs moderately reduces ZIKV replication by approximately one log10 in Human Neural Stem Cells, while also releasing immature virions that have lost their infectivity. This inhibition impairs furin-mediated prM cleavage, ultimately blocking ZIKV maturation. In summary, our study highlights NAAA as a host target for ZIKV infection.


Subject(s)
Zika Virus Infection , Zika Virus , Humans , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/metabolism , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Zika Virus Infection/drug therapy
5.
Elife ; 122023 06 05.
Article in English | MEDLINE | ID: mdl-37272619

ABSTRACT

WDR62 is a spindle pole-associated scaffold protein with pleiotropic functions. Recessive mutations in WDR62 cause structural brain abnormalities and account for the second most common cause of autosomal recessive primary microcephaly (MCPH), indicating WDR62 as a critical hub for human brain development. Here, we investigated WDR62 function in corticogenesis through the analysis of a C-terminal truncating mutation (D955AfsX112). Using induced Pluripotent Stem Cells (iPSCs) obtained from a patient and his unaffected parent, as well as isogenic corrected lines, we generated 2D and 3D models of human neurodevelopment, including neuroepithelial stem cells, cerebro-cortical progenitors, terminally differentiated neurons, and cerebral organoids. We report that WDR62 localizes to the Golgi apparatus during interphase in cultured cells and human fetal brain tissue, and translocates to the mitotic spindle poles in a microtubule-dependent manner. Moreover, we demonstrate that WDR62 dysfunction impairs mitotic progression and results in alterations of the neurogenic trajectories of iPSC neuroderivatives. In summary, impairment of WDR62 localization and function results in severe neurodevelopmental abnormalities, thus delineating new mechanisms in the etiology of MCPH.


Subject(s)
Cell Cycle Proteins , Golgi Apparatus , Microcephaly , Nerve Tissue Proteins , Spindle Poles , Humans , Microcephaly/genetics , Nerve Tissue Proteins/metabolism , Cell Cycle Proteins/metabolism , Male , Induced Pluripotent Stem Cells , Mitosis , Child , Adolescent
6.
Small ; 19(30): e2205871, 2023 07.
Article in English | MEDLINE | ID: mdl-37058009

ABSTRACT

Mechanical stimulation modulates neural development and neuronal activity. In a previous study, magnetic "nano-pulling" is proposed as a tool to generate active forces. By loading neural cells with magnetic nanoparticles (MNPs), a precise force vector is remotely generated through static magnetic fields. In the present study, human neural stem cells (NSCs) are subjected to a standard differentiation protocol, in the presence or absence of nano-pulling. Under mechanical stimulation, an increase in the length of the neural processes which showed an enrichment in microtubules, endoplasmic reticulum, and mitochondria is found. A stimulation lasting up to 82 days induces a strong remodeling at the level of synapse density and a re-organization of the neuronal network, halving the time required for the maturation of neural precursors into neurons. The MNP-loaded NSCs are then transplanted into mouse spinal cord organotypic slices, demonstrating that nano-pulling stimulates the elongation of the NSC processes and modulates their orientation even in an ex vivo model. Thus, it is shown that active mechanical stimuli can guide the outgrowth of NSCs transplanted into the spinal cord tissue. The findings suggest that mechanical forces play an important role in neuronal maturation which could be applied in regenerative medicine.


Subject(s)
Neural Stem Cells , Spinal Cord Injuries , Mice , Animals , Humans , Neurons , Spinal Cord/physiology , Cell Differentiation/physiology , Neurogenesis , Cells, Cultured
7.
Front Neuroanat ; 17: 1130797, 2023.
Article in English | MEDLINE | ID: mdl-36935652

ABSTRACT

The human brain is the most complex structure generated during development. Unveiling the ontogenesis and the intrinsic organization of specific neural networks may represent a key to understanding the physio-pathological aspects of different brain areas. The cortico-thalamic and thalamo-cortical (CT-TC) circuits process and modulate essential tasks such as wakefulness, sleep and memory, and their alterations may result in neurodevelopmental and psychiatric disorders. These pathologies are reported to affect specific neural populations but may also broadly alter physiological connections and thus dysregulate brain network generation, communication, and function. More specifically, the CT-TC system is reported to be severely affected in disorders impacting superior brain functions, such as schizophrenia (SCZ), bipolar disorder, autism spectrum disorders or epilepsy. In this review, the focus will be on CT development, and the models exploited to uncover and comprehend its molecular and cellular mechanisms. In parallel to animal models, still fundamental to unveil human neural network establishment, advanced in vitro platforms, such as brain organoids derived from human pluripotent stem cells, will be discussed. Indeed, organoids and assembloids represent unique tools to study and accelerate fundamental research in CT development and its dysfunctions. We will then discuss recent cutting-edge contributions, including in silico approaches, concerning ontogenesis, specification, and function of the CT-TC circuitry that generates connectivity maps in physiological and pathological conditions.

8.
Stem Cell Reports ; 17(7): 1683-1698, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35714598

ABSTRACT

Congenital alterations in the levels of the transcription factor Forkhead box g1 (FOXG1) coding gene trigger "FOXG1 syndrome," a spectrum that recapitulates birth defects found in the "congenital Zika syndrome," such as microcephaly and other neurodevelopmental conditions. Here, we report that Zika virus (ZIKV) infection alters FOXG1 nuclear localization and causes its downregulation, thus impairing expression of genes involved in cell replication and apoptosis in several cell models, including human neural progenitor cells. Growth factors, such as EGF and FGF2, and Thr271 residue located in FOXG1 AKT domain, take part in the nuclear displacement and apoptosis protection, respectively. Finally, by progressive deletion of FOXG1 sequence, we identify the C-terminus and the residues 428-481 as critical domains. Collectively, our data suggest a causal mechanism by which ZIKV affects FOXG1, its target genes, cell cycle progression, and survival of human neural progenitors, thus contributing to microcephaly.


Subject(s)
Microcephaly , Neural Stem Cells , Zika Virus Infection , Zika Virus , Down-Regulation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Zika Virus/physiology , Zika Virus Infection/genetics
9.
Front Cell Neurosci ; 16: 858347, 2022.
Article in English | MEDLINE | ID: mdl-35573835

ABSTRACT

As microtubule-organizing centers (MTOCs), centrosomes play a pivotal role in cell division, neurodevelopment and neuronal maturation. Among centrosomal proteins, centrin-2 (CETN2) also contributes to DNA repair mechanisms which are fundamental to prevent genomic instability during neural stem cell pool expansion. Nevertheless, the expression profile of CETN2 in human neural stem cells and their progeny is currently unknown. To address this question, we interrogated a platform of human neuroepithelial stem (NES) cells derived from post mortem developing brain or established from pluripotent cells and demonstrated that while CETN2 retains its centrosomal location in proliferating NES cells, its expression pattern changes upon differentiation. In particular, we found that CETN2 is selectively expressed in mature astrocytes with a broad cytoplasmic distribution. We then extended our findings on human autoptic nervous tissue samples. We investigated CETN2 distribution in diverse anatomical areas along the rostro-caudal neuraxis and pointed out a peculiar topography of CETN2-labeled astrocytes in humans which was not appreciable in murine tissues, where CETN2 was mostly confined to ependymal cells. As a prototypical condition with glial overproliferation, we also explored CETN2 expression in glioblastoma multiforme (GBM), reporting a focal concentration of CETN2 in neoplastic astrocytes. This study expands CETN2 localization beyond centrosomes and reveals a unique expression pattern that makes it eligible as a novel astrocytic molecular marker, thus opening new roads to glial biology and human neural conditions.

10.
Sci Rep ; 12(1): 2341, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149717

ABSTRACT

The growth of glioblastoma (GBM), one of the deadliest adult cancers, is fuelled by a subpopulation of stem/progenitor cells, which are thought to be the source of resistance and relapse after treatment. Re-engagement of a latent capacity of these cells to re-enter a trajectory resulting in cell differentiation is a potential new therapeutic approach for this devastating disease. ASCL1, a proneural transcription factor, plays a key role in normal brain development and is also expressed in a subset of GBM cells, but fails to engage a full differentiation programme in this context. Here, we investigated the barriers to ASCL1-driven differentiation in GBM stem cells. We see that ASCL1 is highly phosphorylated in GBM stem cells where its expression is compatible with cell proliferation. However, overexpression of a form of ASCL1 that cannot be phosphorylated on Serine-Proline sites drives GBM cells down a neuronal lineage and out of cell cycle more efficiently than its wild-type counterpart, an effect further enhanced by deletion of the inhibitor of differentiation ID2, indicating mechanisms to reverse the block to GBM cell differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/physiopathology , Glioblastoma/metabolism , Glioblastoma/physiopathology , Inhibitor of Differentiation Protein 2/genetics , Neoplastic Stem Cells/metabolism , Amino Acid Motifs , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Cell Cycle , Cell Differentiation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Inhibitor of Differentiation Protein 2/metabolism , Neoplastic Stem Cells/cytology , Phosphorylation
11.
Prog Mol Biol Transl Sci ; 182: 403-438, 2021.
Article in English | MEDLINE | ID: mdl-34175049

ABSTRACT

The recent advent of genome editing techniques and their rapid improvement paved the way in establishing innovative human neurological disease models and in developing new therapeutic opportunities. Human pluripotent (both induced or naive) stem cells and neural stem cells represent versatile tools to be applied to multiple research needs and, together with genomic snip and fix tools, have recently made possible the creation of unique platforms to directly investigate several human neural affections. In this chapter, we will discuss genome engineering tools, and their recent improvements, applied to the stem cell field, focusing on how these two technologies may be pivotal instruments to deeply unravel molecular mechanisms underlying development and function, as well as disorders, of the human brain. We will review how these frontier technologies may be exploited to investigate or treat severe neurodevelopmental disorders, such as microcephaly, autism spectrum disorder, schizophrenia, as well as neurodegenerative conditions, including Parkinson's disease, Huntington's disease, Alzheimer's disease, and spinal muscular atrophy.


Subject(s)
Autism Spectrum Disorder , Induced Pluripotent Stem Cells , Neural Stem Cells , Neurodegenerative Diseases , Gene Editing , Humans
12.
Front Cell Dev Biol ; 9: 637565, 2021.
Article in English | MEDLINE | ID: mdl-33718375

ABSTRACT

Snap29 is a conserved regulator of membrane fusion essential to complete autophagy and to support other cellular processes, including cell division. In humans, inactivating SNAP29 mutations causes CEDNIK syndrome, a rare multi-systemic disorder characterized by congenital neuro-cutaneous alterations. The fibroblasts of CEDNIK patients show alterations of the Golgi apparatus (GA). However, whether and how Snap29 acts at the GA is unclear. Here we investigate SNAP29 function at the GA and endoplasmic reticulum (ER). As part of the elongated structures in proximity to these membrane compartments, a pool of SNAP29 forms a complex with Syntaxin18, or with Syntaxin5, which we find is required to engage SEC22B-loaded vesicles. Consistent with this, in HeLa cells, in neuroepithelial stem cells, and in vivo, decreased SNAP29 activity alters GA architecture and reduces ER to GA trafficking. Our data reveal a new regulatory function of Snap29 in promoting secretory trafficking.

13.
Cells ; 9(8)2020 08 12.
Article in English | MEDLINE | ID: mdl-32806773

ABSTRACT

Building and functioning of the human brain requires the precise orchestration and execution of myriad molecular and cellular processes, across a multitude of cell types and over an extended period of time. Dysregulation of these processes affects structure and function of the brain and can lead to neurodevelopmental, neurological, or psychiatric disorders. Multiple environmental stimuli affect neural stem cells (NSCs) at several levels, thus impairing the normal human neurodevelopmental program. In this review article, we will delineate the main mechanisms of infection adopted by several neurotropic pathogens, and the selective NSC vulnerability. In particular, TORCH agents, i.e., Toxoplasma gondii, others (including Zika virus and Coxsackie virus), Rubella virus, Cytomegalovirus, and Herpes simplex virus, will be considered for their devastating effects on NSC self-renewal with the consequent neural progenitor depletion, the cellular substrate of microcephaly. Moreover, new evidence suggests that some of these agents may also affect the NSC progeny, producing long-term effects in the neuronal lineage. This is evident in the paradigmatic example of the neurodegeneration occurring in Alzheimer's disease.


Subject(s)
Alzheimer Disease/parasitology , Alzheimer Disease/virology , Microcephaly/parasitology , Microcephaly/virology , Neural Stem Cells/parasitology , Neural Stem Cells/virology , Neurodevelopmental Disorders/parasitology , Neurodevelopmental Disorders/virology , Animals , DNA Virus Infections/complications , DNA Virus Infections/virology , DNA Viruses/pathogenicity , Host-Pathogen Interactions , Humans , Mice , RNA Virus Infections/complications , RNA Virus Infections/virology , RNA Viruses/pathogenicity , Toxoplasma/pathogenicity , Toxoplasmosis/parasitology , Virulence
14.
Cereb Cortex ; 29(5): 2115-2124, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29688344

ABSTRACT

DACH1 is the human homolog of the Drosophila dachshund gene, which is involved in the development of the eye, nervous system, and limbs in the fly. Here, we systematically investigate DACH1 expression patterns during human neurodevelopment, from 5 to 21 postconceptional weeks. By immunodetection analysis, we found that DACH1 is highly expressed in the proliferating neuroprogenitors of the developing cortical ventricular and subventricular regions, while it is absent in the more differentiated cortical plate. Single-cell global transcriptional analysis revealed that DACH1 is specifically enriched in neuroepithelial and ventricular radial glia cells of the developing human neocortex. Moreover, we describe a previously unreported DACH1 expression in the human striatum, in particular in the striatal medium spiny neurons. This finding qualifies DACH1 as a new striatal projection neuron marker, together with PPP1R1B, BCL11B, and EBF1. We finally compared DACH1 expression profile in human and mouse forebrain, where we observed spatio-temporal similarities in its expression pattern thus providing a precise developmental description of DACH1 in the 2 mammalian species.


Subject(s)
Corpus Striatum/embryology , Corpus Striatum/metabolism , Eye Proteins/metabolism , Neocortex/embryology , Neocortex/metabolism , Neuroglia/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Aborted Fetus/embryology , Aborted Fetus/metabolism , Ependymoglial Cells/metabolism , Gestational Age , Humans , Lateral Ventricles/embryology , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Neuroepithelial Cells/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Species Specificity
15.
Nat Commun ; 9(1): 3419, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143638

ABSTRACT

Traumatic spinal cord injury results in persistent disability due to disconnection of surviving neural elements. Neural stem cell transplantation has been proposed as a therapeutic option, but optimal cell type and mechanistic aspects remain poorly defined. Here, we describe robust engraftment into lesioned immunodeficient mice of human neuroepithelial stem cells derived from the developing spinal cord and maintained in self-renewing adherent conditions for long periods. Extensive elongation of both graft and host axons occurs. Improved functional recovery after transplantation depends on neural relay function through the grafted neurons, requires the matching of neural identity to the anatomical site of injury, and is accompanied by expression of specific marker proteins. Thus, human neuroepithelial stem cells may provide an anatomically specific relay function for spinal cord injury recovery.


Subject(s)
Neural Stem Cells/cytology , Spinal Cord Regeneration/physiology , Animals , Axons/metabolism , Cell Differentiation/physiology , Cell Line , Cell Survival/physiology , Cells, Cultured , Female , Humans , Male , Mice , Neural Stem Cells/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Stem Cell Transplantation
16.
Science ; 358(6366): 1027-1032, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29170230

ABSTRACT

To better understand the molecular and cellular differences in brain organization between human and nonhuman primates, we performed transcriptome sequencing of 16 regions of adult human, chimpanzee, and macaque brains. Integration with human single-cell transcriptomic data revealed global, regional, and cell-type-specific species expression differences in genes representing distinct functional categories. We validated and further characterized the human specificity of genes enriched in distinct cell types through histological and functional analyses, including rare subpallial-derived interneurons expressing dopamine biosynthesis genes enriched in the human striatum and absent in the nonhuman African ape neocortex. Our integrated analysis of the generated data revealed diverse molecular and cellular features of the phylogenetic reorganization of the human brain across multiple levels, with relevance for brain function and disease.


Subject(s)
Macaca/genetics , Neocortex/growth & development , Neocortex/metabolism , Neural Pathways/metabolism , Pan troglodytes/genetics , Transcriptome , Animals , Gene Expression Profiling , Humans , Interneurons/metabolism , Phylogeny , Species Specificity
17.
Cell Rep ; 16(10): 2576-2592, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568284

ABSTRACT

The mechanisms underlying Zika virus (ZIKV)-related microcephaly and other neurodevelopment defects remain poorly understood. Here, we describe the derivation and characterization, including single-cell RNA-seq, of neocortical and spinal cord neuroepithelial stem (NES) cells to model early human neurodevelopment and ZIKV-related neuropathogenesis. By analyzing human NES cells, organotypic fetal brain slices, and a ZIKV-infected micrencephalic brain, we show that ZIKV infects both neocortical and spinal NES cells as well as their fetal homolog, radial glial cells (RGCs), causing disrupted mitoses, supernumerary centrosomes, structural disorganization, and cell death. ZIKV infection of NES cells and RGCs causes centrosomal depletion and mitochondrial sequestration of phospho-TBK1 during mitosis. We also found that nucleoside analogs inhibit ZIKV replication in NES cells, protecting them from ZIKV-induced pTBK1 relocalization and cell death. We established a model system of human neural stem cells to reveal cellular and molecular mechanisms underlying neurodevelopmental defects associated with ZIKV infection and its potential treatment.


Subject(s)
Mitosis , Neural Stem Cells/enzymology , Neural Stem Cells/virology , Neuroepithelial Cells/virology , Neuroglia/virology , Protein Serine-Threonine Kinases/metabolism , Zika Virus/pathogenicity , Brain/embryology , Brain/pathology , Brain/virology , Cell Death/drug effects , Centrosome/drug effects , Centrosome/metabolism , Fetus/virology , Gene Expression Profiling , Humans , Immunity, Innate/drug effects , Microcephaly/pathology , Microcephaly/virology , Mitochondria/drug effects , Mitochondria/metabolism , Mitosis/drug effects , Neocortex/pathology , Neural Stem Cells/immunology , Neural Stem Cells/ultrastructure , Neuroepithelial Cells/drug effects , Neuroepithelial Cells/immunology , Neuroepithelial Cells/ultrastructure , Neuroglia/pathology , Neuroglia/ultrastructure , Neurons/drug effects , Neurons/pathology , Neurons/virology , Neuroprotective Agents/pharmacology , Nucleosides/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Spinal Cord/pathology , Transcription, Genetic/drug effects , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus/physiology , Zika Virus/ultrastructure , Zika Virus Infection/pathology , Zika Virus Infection/virology , Axl Receptor Tyrosine Kinase
19.
Dev Biol ; 411(1): 25-37, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26806704

ABSTRACT

HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells.


Subject(s)
Cell Differentiation/genetics , Epithelial-Mesenchymal Transition/genetics , HMGA2 Protein/physiology , Neural Crest/embryology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Animals , Cell Line, Tumor , Cell Movement/genetics , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks/genetics , HMGA2 Protein/genetics , MSX1 Transcription Factor/genetics , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Morpholinos/genetics , Neural Crest/cytology , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/metabolism , Xenopus Proteins/genetics
20.
Nat Neurosci ; 17(12): 1804-15, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25383901

ABSTRACT

The complexity of the human brain derives from the intricate interplay of molecular instructions during development. Here we systematically investigated gene expression changes in the prenatal human striatum and cerebral cortex during development from post-conception weeks 2 to 20. We identified tissue-specific gene coexpression networks, differentially expressed genes and a minimal set of bimodal genes, including those encoding transcription factors, that distinguished striatal from neocortical identities. Unexpected differences from mouse striatal development were discovered. We monitored 36 determinants at the protein level, revealing regional domains of expression and their refinement, during striatal development. We electrophysiologically profiled human striatal neurons differentiated in vitro and determined their refined molecular and functional properties. These results provide a resource and opportunity to gain global understanding of how transcriptional and functional processes converge to specify human striatal and neocortical neurons during development.


Subject(s)
Corpus Striatum/embryology , Corpus Striatum/physiology , Fetal Development/physiology , Gene Regulatory Networks/physiology , Action Potentials/physiology , Cell Differentiation/physiology , Cells, Cultured , HEK293 Cells , Humans , Organ Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...