Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Mol Cell ; 82(15): 2779-2796.e10, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35675814

RESUMEN

Despite a long appreciation for the role of nonsense-mediated mRNA decay (NMD) in destroying faulty, disease-causing mRNAs and maintaining normal, physiologic mRNA abundance, additional effectors that regulate NMD activity in mammalian cells continue to be identified. Here, we describe a haploid-cell genetic screen for NMD effectors that has unexpectedly identified 13 proteins constituting the AKT signaling pathway. We show that AKT supersedes UPF2 in exon-junction complexes (EJCs) that are devoid of RNPS1 but contain CASC3, defining an unanticipated insulin-stimulated EJC. Without altering UPF1 RNA binding or ATPase activity, AKT-mediated phosphorylation of the UPF1 CH domain at T151 augments UPF1 helicase activity, which is critical for NMD and also decreases the dependence of helicase activity on ATP. We demonstrate that upregulation of AKT signaling contributes to the hyperactivation of NMD that typifies Fragile X syndrome, as exemplified using FMR1-KO neural stem cells derived from induced pluripotent stem cells.


Asunto(s)
Degradación de ARNm Mediada por Codón sin Sentido , Proteínas Proto-Oncogénicas c-akt , Animales , Codón sin Sentido/genética , Exones/genética , Mamíferos/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Helicasas/genética , ARN Helicasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
2.
Mol Cell Neurosci ; 123: 103770, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36055520

RESUMEN

Many neurodegenerative diseases have a multifactorial etiology and variable course of progression that cannot be explained by current models. Neurotropic viruses have long been suggested to play a role in these diseases, although their exact contributions remain unclear. Human herpesvirus 6A (HHV-6A) is one of the most common viruses detected in the adult brain, and has been clinically associated with multiple sclerosis (MS), and, more recently, Alzheimer's disease (AD). HHV-6A is a ubiquitous viral pathogen capable of infecting glia and neurons. Primary infection in childhood is followed by the induction of latency, characterized by expression of the U94A viral transcript in the absence of viral replication. Here we examine the effects of U94A on cells of the central nervous system. We found that U94A expression inhibits the migration and impairs cytoplasmic maturation of human oligodendrocyte precursor cells (OPCs) without affecting their viability, a phenotype that may contribute to the failure of remyelination seen in many patients with MS. A subsequent proteomics analysis of U94A expression OPCs revealed altered expression of genes involved in tubulin associated cytoskeletal regulation. As HHV-6A seems to significantly be associated with early AD pathology, we extended our initially analysis of the impact of U94A on human derived neurons. We found that U94A expression inhibits neurite outgrowth of primary human cortical neurons and impairs synapse maturation. Based on these data we suggest that U94A expression by latent HHV-6A in glial cells and neurons renders them susceptible to dysfunction and degeneration. Therefore, latent viral infections of the brain represent a unique pathological risk factor that may contribute to disease processes.


Asunto(s)
Herpesvirus Humano 6 , Esclerosis Múltiple , Células Precursoras de Oligodendrocitos , Humanos , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/metabolismo , Sistema Nervioso Central , Neuroglía
3.
PLoS Biol ; 14(12): e1002583, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27977664

RESUMEN

Neurodegenerative lysosomal storage disorders (LSDs) are severe and untreatable, and mechanisms underlying cellular dysfunction are poorly understood. We found that toxic lipids relevant to three different LSDs disrupt multiple lysosomal and other cellular functions. Unbiased drug discovery revealed several structurally distinct protective compounds, approved for other uses, that prevent lysosomal and cellular toxicities of these lipids. Toxic lipids and protective agents show unexpected convergence on control of lysosomal pH and re-acidification as a critical component of toxicity and protection. In twitcher mice (a model of Krabbe disease [KD]), a central nervous system (CNS)-penetrant protective agent rescued myelin and oligodendrocyte (OL) progenitors, improved motor behavior, and extended lifespan. Our studies reveal shared principles relevant to several LSDs, in which diverse cellular and biochemical disruptions appear to be secondary to disruption of lysosomal pH regulation by specific lipids. These studies also provide novel protective strategies that confer therapeutic benefits in a mouse model of a severe LSD.


Asunto(s)
Ácidos/metabolismo , Modelos Animales de Enfermedad , Enfermedades por Almacenamiento Lisosomal/metabolismo , Lisosomas/metabolismo , Esfingolípidos/metabolismo , Animales , Colforsina/farmacología , Humanos , Ratones , Células Madre/citología
4.
Mol Cell Neurosci ; 92: 128-136, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29969654

RESUMEN

Astrocytes play an indispensable role in maintaining a healthy, functional neural network in the central nervous system (CNS). A primary function of CNS astrocytes is to support the survival and function of neurons. In response to injury, astrocytes take on a reactive phenotype, which alters their molecular functions. Reactive astrocytes have been reported to be both beneficial and harmful to the CNS recovery process subsequent to injury. Understanding the molecular processes and regulatory proteins that determine the extent to which an astrocyte hinders or supports neuronal survival is important within the context of CNS repair. One protein that plays a role in modulating cellular survival is transglutaminase 2 (TG2). Global deletion of TG2 results in beneficial outcomes subsequent to in vivo ischemic brain injury. Ex vivo studies have also implicated TG2 as a negative regulator of astrocyte viability subsequent to injury. In this study we show that knocking down TG2 in astrocytes significantly increases their ability to protect neurons from oxygen glucose deprivation (OGD)/reperfusion injury. To begin to understand how deletion of TG2 in astrocytes improves their ability to protect neurons from injury, we performed transcriptome analysis of wild type and TG2-/- astrocytes. TG2 deletion resulted in alterations in genes involved in extracellular matrix remodeling, cell adhesion and axon growth/guidance. In addition, the majority of genes that showed increases in the TG2-/- astrocytes had predicted cJun/AP-1 binding motifs in their promoters. Furthermore, phospho-cJun levels were robustly elevated in TG2-/- astrocytes, a finding which was consistent with the increase in expression of AP-1 responsive genes. These in vitro data were subsequently extended into an in vivo model to determine whether the absence of astrocytic TG2 improves outcomes after CNS injury. Our results show that, following a spinal cord injury, scar formation is significantly attenuated in mice with astrocyte-specific TG2 deletion compared to mice expressing normal TG2 levels. Taken together, these data indicate that TG2 plays a pivotal role in mediating reactive astrocyte properties following CNS injury. Further, the data suggest that limiting the AP-1 mediated pro-survival injury response may be a contributing factor to that the detrimental effects of astrocytic TG2.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Unión al GTP/genética , Regeneración Nerviosa , Traumatismos de la Médula Espinal/metabolismo , Transglutaminasas/genética , Animales , Orientación del Axón , Hipoxia de la Célula , Células Cultivadas , Proteínas de Unión al GTP/metabolismo , Glucosa/deficiencia , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Traumatismos de la Médula Espinal/genética , Factor de Transcripción AP-1/metabolismo , Transcriptoma , Transglutaminasas/metabolismo
5.
Hum Mol Genet ; 24(22): 6331-49, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26310626

RESUMEN

Ataxia-telangiectasia (A-T) is a rare multi-system disorder caused by mutations in the ATM gene. Significant heterogeneity exists in the underlying genetic mutations and clinical phenotypes. A number of mouse models have been generated that harbor mutations in the distal region of the gene, and a recent study suggests the presence of residual ATM protein in the brain of one such model. These mice recapitulate many of the characteristics of A-T seen in humans, with the notable exception of neurodegeneration. In order to study how an N-terminal mutation affects the disease phenotype, we generated an inducible Atm mutant mouse model (Atm(tm1Mmpl/tm1Mmpl), referred to as A-T [M]) predicted to express only the first 62 amino acids of Atm. Cells derived from A-T [M] mutant mice exhibited reduced cellular proliferation and an altered DNA damage response, but surprisingly, showed no evidence of an oxidative imbalance. Examination of the A-T [M] animals revealed an altered immunophenotype consistent with A-T. In contrast to mice harboring C-terminal Atm mutations that disproportionately develop thymic lymphomas, A-T [M] mice developed lymphoma at a similar rate as human A-T patients. Morphological analyses of A-T [M] cerebella revealed no substantial cellular defects, similar to other models of A-T, although mice display behavioral defects consistent with cerebellar dysfunction. Overall, these results suggest that loss of Atm is not necessarily associated with an oxidized phenotype as has been previously proposed and that loss of ATM protein is not sufficient to induce cerebellar degeneration in mice.


Asunto(s)
Ataxia Telangiectasia/genética , Linfoma de Células T/genética , Mutación , Animales , Ataxia Telangiectasia/enzimología , Ataxia Telangiectasia/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Conducta Animal/fisiología , Proteínas de Ciclo Celular/genética , Daño del ADN , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Estudios de Asociación Genética , Humanos , Incidencia , Linfoma de Células T/enzimología , Linfoma de Células T/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Proteínas Supresoras de Tumor/genética
6.
Glia ; 64(2): 227-39, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26469940

RESUMEN

Astroglial dysfunction plays an important role in neurodegenerative diseases otherwise attributed to neuronal loss of function. Here we focus on the role of astroglia in ataxia-telangiectasia (A-T), a disease caused by mutations in the ataxia-telangiectasia mutated (ATM) gene. A hallmark of A-T pathology is progressive loss of cerebellar neurons, but the mechanisms that impact neuronal survival are unclear. We now provide a possible mechanism by which A-T astroglia affect the survival of cerebellar neurons. As astroglial functions are difficult to study in an in vivo setting, particularly in the cerebellum where these cells are intertwined with the far more numerous neurons, we conducted in vitro coculture experiments that allow for the generation and pharmacological manipulation of purified cell populations. Our analyses revealed that cerebellar astroglia isolated from Atm mutant mice show decreased expression of the cystine/glutamate exchanger subunit xCT, glutathione (GSH) reductase, and glutathione-S-transferase. We also found decreased levels of intercellular and secreted GSH in A-T astroglia. Metabolic labeling of l-cystine, the major precursor for GSH, revealed that a key component of the defect in A-T astroglia is an impaired ability to import this rate-limiting precursor for the production of GSH. This impairment resulted in suboptimal extracellular GSH supply, which in turn impaired survival of cerebellar neurons. We show that by circumventing the xCT-dependent import of L-cystine through addition of N-acetyl-L-cysteine (NAC) as an alternative cysteine source, we were able to restore GSH levels in A-T mutant astroglia providing a possible future avenue for targeted therapeutic intervention.


Asunto(s)
Astrocitos/metabolismo , Cerebelo/metabolismo , Glutatión/metabolismo , Homeostasis/fisiología , Acetilcisteína/metabolismo , Adolescente , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Supervivencia Celular/fisiología , Técnicas de Cocultivo , Cistina/metabolismo , Espacio Extracelular/metabolismo , Glutatión Reductasa/metabolismo , Humanos , Espacio Intracelular/metabolismo , Ratones de la Cepa 129 , Ratones Transgénicos , Mutación , Neuronas/fisiología
7.
J Neurosci ; 34(7): 2438-43, 2014 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24523534

RESUMEN

Traumatic spinal cord injury (SCI) results in a cascade of tissue responses leading to cell death, axonal degeneration, and glial scar formation, exacerbating the already hostile environment and further inhibiting axon regeneration. Overcoming these inhibitory cues and promoting axonal regeneration is one of the primary targets in developing a cure for SCI. Previously, we demonstrated that transplantation of bone morphogenetic protein (BMP)-induced astrocytes derived from embryonic glial-restricted precursors (GDAs(BMP)) promotes extensive axonal growth and motor function recovery in a rodent spinal cord injury model. Here, we identify periostin (POSTN), a secreted protein, as a key component of GDA(BMP)-induced axonal regeneration. POSTN is highly expressed by GDAs(BMP) and the perturbation of POSTN expression by shRNA diminished GDA(BMP)-induced neurite extension in vitro. We also found that recombinant POSTN is sufficient to overcome the inhibitory effect of scar-associated molecules and promote neurite extension in vitro by signaling through focal adhesion kinase and Akt. Furthermore, transplantation of POSTN-deficient GDAs(BMP) into the injured rat spinal cord resulted in compromised axonal regeneration, indicating that POSTN plays an essential role in GDA(BMP)-mediated axonal regeneration. This finding reveals not only one of the major mechanisms underlying GDA(BMP)-dependent recovery from SCI, but also the potential of POSTN as a therapeutic agent for traumatic injury of the CNS.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/trasplante , Moléculas de Adhesión Celular/metabolismo , Regeneración Nerviosa/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Axones/metabolismo , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células-Madre Neurales/citología , Ratas , Ratas Sprague-Dawley
8.
Nat Med ; 11(3): 277-83, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15723074

RESUMEN

Vanishing white matter disease (VWM) is a heritable leukodystrophy linked to mutations in translation initiation factor 2B (eIF2B). Although the clinical course of this disease has been relatively well described, the cellular consequences of EIF2B mutations on neural cells are unknown. Here we have established cell cultures from the brain of an individual with VWM carrying mutations in subunit 5 of eIF2B (encoded by EIF2B5). Despite the extensive demyelination apparent in this VWM patient, normal-appearing oligodendrocytes were readily generated in vitro. In contrast, few GFAP-expressing (GFAP+) astrocytes were present in primary cultures, induction of astrocytes was severely compromised, and the few astrocytes generated showed abnormal morphologies and antigenic phenotypes. Lesions in vivo also lacked GFAP+ astrocytes. RNAi targeting of EIF2B5 severely compromised the induction of GFAP+ cells from normal human glial progenitors. This raises the possibility that a deficiency in astrocyte function may contribute to the loss of white matter in VWM leukodystrophy.


Asunto(s)
Astrocitos/citología , Encefalopatías/genética , Factor 2B Eucariótico de Iniciación/genética , Proteína Ácida Fibrilar de la Glía/biosíntesis , Enfermedades Neurodegenerativas/genética , Secuencia de Bases , Encefalopatías/fisiopatología , Diferenciación Celular , Células Cultivadas , Niño , Enfermedades Desmielinizantes/patología , Factor 2B Eucariótico de Iniciación/fisiología , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Mutación , Enfermedades Neurodegenerativas/fisiopatología
9.
Osteoarthr Cartil Open ; 4(1): 100227, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36474470

RESUMEN

Objective: The objective of this study is to understand the role of altered in vivo mechanical environments in knee joints post anterior cruciate ligament (ACL)-injury in chondrocyte vulnerability against mechanical stimuli and in the progression of post-traumatic osteoarthritis (PT-OA). Methods: Differential in vivo mechanical environments were induced by unilateral ACL-injury (uni-ACL-I) and bilateral ACL-injury (bi-ACL-I) in 8-week-old female C57BL/6 mice. The gait parameters, the mechano-vulnerability of in situ chondrocytes, Young's moduli of cartilage extracellular matrix (ECM), and the histological assessment of OA severity (OARSI score) were compared between control and experimental groups at 0∼8-weeks post-ACL-injury. Results: We found that bi-ACL-I mice experience higher joint-loading on their both injured limbs, but uni-ACL-I mice balance their joint-loading between injured and uninjured hind limbs resulting in a reduced joint-loading during gait. We also found that at 4- and 8-week post-injury the higher weight-bearing hind limbs (i.e., bi-ACL-I) had the increased area of chondrocyte death induced by impact loading and higher OARSI score than the lower weight-bearing limbs (uni-ACL-I). Additionally, we found that at 8-weeks post-injury the ECM became stiffer in bi-ACL-I joints and softer in uni-ACL-I joints. Conclusions: Our results show that ACL-injured limbs with lower in vivo joint-loading develops PT-OA significantly slower than injured limbs with higher joint-loading during gait. Our data also indicate that articular chondrocytes in severe PT-OA are more fragile from mechanical impacts than chondrocytes in healthy or mild PT-OA. Thus, preserving physiologic joint-loads on injured joints will reduce chondrocyte death post-injury and may delay PT-OA progression.

10.
Curr Opin Neurol ; 24(6): 570-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22027545

RESUMEN

PURPOSE OF REVIEW: Central to the obstacles to be overcome in moving promising cell-based therapies from the laboratory to the clinic is that of determining which of the many cell types being examined are optimal for repairing particular lesions. RECENT FINDINGS: Our studies on astrocyte replacement therapies demonstrate clearly that some cells are far better than others at promoting recovery in spinal cord injury and that, at least in some cases, transplanting undifferentiated precursor cells is far less useful than transplanting specific astrocytes derived from those precursor cells. But further comparison between different approaches is hindered by the difficulties in replicating results between laboratories, even for well defined pharmacological agents and bioactive proteins. These difficulties in replication appear most likely to be due to unrecognized nuances in lesion characteristics and in the details of delivery of therapies. SUMMARY: We propose that the challenge of reproducibility provides a critical opportunity for refining cell-based therapies. If the utility of a particular approach is so restricted that even small changes in lesions or treatment protocols eliminate benefit, then the variability inherent in clinical injuries will frustrate translation. In contrast, rising to this challenge may enable discovery of refinements needed to confer the robustness needed for successful clinical trials.


Asunto(s)
Trasplante de Células/métodos , Enfermedades del Sistema Nervioso Central/terapia , Sistema Nervioso Central/patología , Animales , Astrocitos/trasplante , Enfermedades del Sistema Nervioso Central/patología , Enfermedades del Sistema Nervioso Central/fisiopatología , Ensayos Clínicos como Asunto , Humanos , Reproducibilidad de los Resultados , Traumatismos de la Médula Espinal/terapia , Resultado del Tratamiento
11.
Nat Cell Biol ; 23(1): 40-48, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33420492

RESUMEN

Loss of the fragile X protein FMRP is a leading cause of intellectual disability and autism1,2, but the underlying mechanism remains poorly understood. We report that FMRP deficiency results in hyperactivated nonsense-mediated mRNA decay (NMD)3,4 in human SH-SY5Y neuroblastoma cells and fragile X syndrome (FXS) fibroblast-derived induced pluripotent stem cells (iPSCs). We examined the underlying mechanism and found that the key NMD factor UPF1 binds directly to FMRP, promoting FMRP binding to NMD targets. Our data indicate that FMRP acts as an NMD repressor. In the absence of FMRP, NMD targets are relieved from FMRP-mediated translational repression so that their half-lives are decreased and, for those NMD targets encoding NMD factors, increased translation produces abnormally high factor levels despite their hyperactivated NMD. Transcriptome-wide alterations caused by NMD hyperactivation have a role in the FXS phenotype. Consistent with this, small-molecule-mediated inhibition of hyperactivated NMD, which typifies iPSCs derived from patients with FXS, restores a number of neurodifferentiation markers, including those not deriving from NMD targets. Our mechanistic studies reveal that many molecular abnormalities in FMRP-deficient cells are attributable-either directly or indirectly-to misregulated NMD.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Eliminación de Gen , Neuroblastoma/patología , Degradación de ARNm Mediada por Codón sin Sentido , Transcriptoma , Estudios de Casos y Controles , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuronas/metabolismo , Neuronas/patología , RNA-Seq , Transactivadores
12.
Genome Biol ; 22(1): 317, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34784943

RESUMEN

BACKGROUND: Fragile X syndrome (FXS) is an intellectual disability attributable to loss of fragile X protein (FMRP). We previously demonstrated that FMRP binds mRNAs targeted for nonsense-mediated mRNA decay (NMD) and that FMRP loss results in hyperactivated NMD and inhibition of neuronal differentiation in human stem cells. RESULTS: We show here that NMD is hyperactivated during the development of the cerebral cortex, hippocampus, and cerebellum in the Fmr1-knockout (KO) mouse during embryonic and early postnatal periods. Our findings demonstrate that NMD regulates many neuronal mRNAs that are important for mouse brain development. CONCLUSIONS: We reveal the abnormal regulation of these mRNAs in the Fmr1-KO mouse, a model of FXS, and highlight the importance of early intervention.


Asunto(s)
Encefalopatías/genética , Encéfalo/crecimiento & desarrollo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Degradación de ARNm Mediada por Codón sin Sentido/genética , Animales , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo
13.
Cells ; 10(11)2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34831164

RESUMEN

Following CNS injury, astrocytes become "reactive" and exhibit pro-regenerative or harmful properties. However, the molecular mechanisms that cause astrocytes to adopt either phenotype are not well understood. Transglutaminase 2 (TG2) plays a key role in regulating the response of astrocytes to insults. Here, we used mice in which TG2 was specifically deleted in astrocytes (Gfap-Cre+/- TG2fl/fl, referred to here as TG2-A-cKO) in a spinal cord contusion injury (SCI) model. Deletion of TG2 from astrocytes resulted in a significant improvement in motor function following SCI. GFAP and NG2 immunoreactivity, as well as number of SOX9 positive cells, were significantly reduced in TG2-A-cKO mice. RNA-seq analysis of spinal cords from TG2-A-cKO and control mice 3 days post-injury identified thirty-seven differentially expressed genes, all of which were increased in TG2-A-cKO mice. Pathway analysis revealed a prevalence for fatty acid metabolism, lipid storage and energy pathways, which play essential roles in neuron-astrocyte metabolic coupling. Excitingly, treatment of wild type mice with the selective TG2 inhibitor VA4 significantly improved functional recovery after SCI, similar to what was observed using the genetic model. These findings indicate the use of TG2 inhibitors as a novel strategy for the treatment of SCI and other CNS injuries.


Asunto(s)
Astrocitos/enzimología , Eliminación de Gen , Proteína Glutamina Gamma Glutamiltransferasa 2/antagonistas & inhibidores , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/complicaciones , Gliosis/patología , Ratones Noqueados , Proteína Glutamina Gamma Glutamiltransferasa 2/metabolismo , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Elife ; 92020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32924936

RESUMEN

The ability of a well-known component of the complement cascade to bind to a variety of receptors has implications for signaling biology, spinal cord injury and, possibly, the evolution of the complement system.


Asunto(s)
Complemento C1q , Células-Madre Neurales , Glicoproteínas de Membrana , Receptores de Complemento
15.
Sci Rep ; 7(1): 3978, 2017 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-28638124

RESUMEN

Progression of demyelinating diseases is caused by an imbalance of two opposing processes: persistent destruction of myelin and myelin repair by differentiating oligodendrocyte progenitor cells (OPCs). Repair that cannot keep pace with destruction results in progressive loss of myelin. Viral infections have long been suspected to be involved in these processes but their specific role remains elusive. Here we describe a novel mechanism by which HHV-6A, a member of the human herpesvirus family, may contribute to inadequate myelin repair after injury.


Asunto(s)
Movimiento Celular , Herpesvirus Humano 6/metabolismo , Células Precursoras de Oligodendrocitos/virología , Proteínas Virales/metabolismo , Latencia del Virus , Células Cultivadas , Enfermedades Desmielinizantes/virología , Humanos , Células Precursoras de Oligodendrocitos/metabolismo
16.
Eur J Hum Genet ; 25(2): 216-221, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27966542

RESUMEN

SIK1 syndrome is a newly described developmental epilepsy disorder caused by heterozygous mutations in the salt-inducible kinase SIK1. To better understand the pathophysiology of SIK1 syndrome, we studied the effects of SIK1 pathogenic sequence variations in human neurons. Primary human fetal cortical neurons were transfected with a lentiviral vector to overexpress wild-type and mutant SIK1 protein. We evaluated the transcriptional activity of known downstream gene targets in neurons expressing mutant SIK1 compared with wild type. We then assayed neuronal morphology by measuring neurite length, number and branching. Truncating SIK1 sequence variations were associated with abnormal MEF2C transcriptional activity and decreased MEF2C protein levels. Epilepsy-causing SIK1 sequence variations were associated with significantly decreased expression of ARC (activity-regulated cytoskeletal-associated) and other synaptic activity response element genes. Assay of mRNA levels for other MEF2C target genes NR4A1 (Nur77) and NRG1, found significantly, decreased the expression of these genes as well. The missense p.(Pro287Thr) SIK1 sequence variation was associated with abnormal neuronal morphology, with significant decreases in mean neurite length, mean number of neurites and a significant increase in proximal branches compared with wild type. Epilepsy-causing SIK1 sequence variations resulted in abnormalities in the MEF2C-ARC pathway of neuronal development and synapse activity response. This work provides the first insights into the mechanisms of pathogenesis in SIK1 syndrome, and extends the ARX-MEF2C pathway in the pathogenesis of developmental epilepsy.


Asunto(s)
Epilepsia/genética , Mutación , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transmisión Sináptica , Células Cultivadas , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Epilepsia/metabolismo , Epilepsia/patología , Células HEK293 , Humanos , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neurregulina-1/genética , Neurregulina-1/metabolismo , Neuronas/patología , Neuronas/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
17.
J Neurosci ; 22(1): 248-56, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11756508

RESUMEN

We have found that the tripotential glial-restricted precursor (GRP) cell of the embryonic rat spinal cord can give rise in vitro to bipotential cells that express defining characteristics of oligodendrocyte-type-2 astrocyte progenitor cells (O2A/OPCs). Generation of O2A/OPCs is regulated by environmental signals and is promoted by platelet-derived growth factor (PDGF), thyroid hormone (TH) and astrocyte-conditioned medium. In contrast to multiple observations indicating that oligodendrocyte precursor cells in the embryonic day 14 (E14) spinal cord are ventrally restricted, GRP cells are already present in both the dorsal and ventral spinal cord at E13.5. Ventral-derived GRP cells, however, were more likely to generate O2A/OPCs and/or oligodendrocytes than were their dorsal counterparts when exposed to TH, PDGF, or even bone morphogenetic protein-4. The simplest explanation of our results is that oligodendrocyte generation occurs as a result of generation of GRP cells from totipotent neuroepithelial stem cells, of O2A/OPCs from GRP cells and, finally, of oligodendrocytes from O2A/OPCs. In this respect, the responsiveness of GRP cells to modulators of this process may represent a central control point in the initiation of this critical developmental sequence. Our findings provide an integration between the earliest known glial precursors and the well-studied O2A/OPCs while opening up new questions concerning the intricate spatial and temporal regulation of precursor cell differentiation in the CNS.


Asunto(s)
Tipificación del Cuerpo , Neuroglía/citología , Médula Espinal/citología , Médula Espinal/embriología , Células Madre/citología , Animales , Antígenos de Diferenciación/biosíntesis , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Proteína Morfogenética Ósea 4 , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/fisiología , Linaje de la Célula/efectos de los fármacos , Células Cultivadas , Células Clonales/citología , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Hormonas Tiroideas/farmacología
18.
Free Radic Biol Med ; 79: 300-23, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25481740

RESUMEN

This review discusses a unique discovery path starting with novel findings on redox regulation of precursor cell and signaling pathway function and identification of a new mechanism by which relatively small changes in redox status can control entire signaling networks that regulate self-renewal, differentiation, and survival. The pathway central to this work, the redox/Fyn/c-Cbl (RFC) pathway, converts small increases in oxidative status to pan-activation of the c-Cbl ubiquitin ligase, which controls multiple receptors and other proteins of central importance in precursor cell and cancer cell function. Integration of work on the RFC pathway with attempts to understand how treatment with systemic chemotherapy causes neurological problems led to the discovery that glioblastomas (GBMs) and basal-like breast cancers (BLBCs) inhibit c-Cbl function through altered utilization of the cytoskeletal regulators Cool-1/ßpix and Cdc42, respectively. Inhibition of these proteins to restore normal c-Cbl function suppresses cancer cell division, increases sensitivity to chemotherapy, disrupts tumor-initiating cell (TIC) activity in GBMs and BLBCs, controls multiple critical TIC regulators, and also allows targeting of non-TICs. Moreover, these manipulations do not increase chemosensitivity or suppress division of nontransformed cells. Restoration of normal c-Cbl function also allows more effective harnessing of estrogen receptor-α (ERα)-independent activities of tamoxifen to activate the RFC pathway and target ERα-negative cancer cells. Our work thus provides a discovery strategy that reveals mechanisms and therapeutic targets that cannot be deduced by standard genetics analyses, which fail to reveal the metabolic information, isoform shifts, protein activation, protein complexes, and protein degradation critical to our discoveries.


Asunto(s)
Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Animales , Humanos , Neoplasias/patología , Neoplasias/terapia , Oxidación-Reducción
19.
EMBO Mol Med ; 6(4): 504-18, 2014 04.
Artículo en Inglés | MEDLINE | ID: mdl-24477866

RESUMEN

In addition to dopaminergic neuron loss, it is clear that Parkinson disease includes other pathological changes, including loss of additional neuronal populations. As a means of addressing multiple pathological changes with a single therapeutically-relevant approach, we employed delayed transplantation of a unique class of astrocytes, GDAs(BMP), that are generated in vitro by directed differentiation of glial precursors. GDAs(BMP) produce multiple agents of interest as treatments for PD and other neurodegenerative disorders, including BDNF, GDNF, neurturin and IGF1. GDAs(BMP) also exhibit increased levels of antioxidant pathway components, including levels of NADPH and glutathione. Delayed GDA(BMP) transplantation into the 6-hydroxydopamine lesioned rat striatum restored tyrosine hydroxylase expression and promoted behavioral recovery. GDA(BMP) transplantation also rescued pathological changes not prevented in other studies, such as the rescue of parvalbumin(+) GABAergic interneurons. Consistent with expression of the synaptic modulatory proteins thrombospondin-1 and 2 by GDAs(BMP), increased expression of the synaptic protein synaptophysin was also observed. Thus, GDAs(BMP) offer a multimodal support cell therapy that provides multiple benefits without requiring prior genetic manipulation.


Asunto(s)
Astrocitos/trasplante , Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedad de Parkinson/terapia , Animales , Astrocitos/citología , Humanos , Masculino , Neuroglía/citología , Neuroglía/trasplante , Ratas , Ratas Endogámicas F344
20.
Cell Stem Cell ; 12(5): 503-4, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23642358

RESUMEN

Generating patient-specific oligodendrocyte progenitors capable of repairing myelination defects observed in multiple neurological afflictions holds great therapeutic potential. Recently in Nature Biotechnology, Najm et al. (2013) and Yang et al. (2013) generated these progenitors by direct reprogramming, bringing us closer to their use in disease analysis and autologous transplantation strategies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA