Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 1923, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772011

RESUMEN

Chronic demyelination in the human CNS is characterized by an inhibitory microenvironment that impairs recruitment and differentiation of oligodendrocyte progenitor cells (OPCs) leading to failed remyelination and axonal atrophy. By network-based transcriptomics, we identified sulfatase 2 (Sulf2) mRNA in activated human primary OPCs. Sulf2, an extracellular endosulfatase, modulates the signaling microenvironment by editing the pattern of sulfation on heparan sulfate proteoglycans. We found that Sulf2 was increased in demyelinating lesions in multiple sclerosis and was actively secreted by human OPCs. In experimental demyelination, elevated OPC Sulf1/2 expression directly impaired progenitor recruitment and subsequent generation of oligodendrocytes thereby limiting remyelination. Sulf1/2 potentiates the inhibitory microenvironment by promoting BMP and WNT signaling in OPCs. Importantly, pharmacological sulfatase inhibition using PI-88 accelerated oligodendrocyte recruitment and remyelination by blocking OPC-expressed sulfatases. Our findings define an important inhibitory role of Sulf1/2 and highlight the potential for modulation of the heparanome in the treatment of chronic demyelinating disease.


Asunto(s)
Diferenciación Celular/genética , Microambiente Celular/genética , Enfermedades Desmielinizantes/genética , Perfilación de la Expresión Génica/métodos , Células Precursoras de Oligodendrocitos/metabolismo , Remielinización/genética , Animales , Axones/metabolismo , Células Cultivadas , Enfermedades Desmielinizantes/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Células Precursoras de Oligodendrocitos/citología , Sulfatasas/genética , Sulfatasas/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
2.
Cell Rep ; 25(12): 3435-3450.e6, 2018 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-30566868

RESUMEN

Human oligodendrocyte progenitor cells (hOPCs) persist into adulthood as an abundant precursor population capable of division and differentiation. The transcriptional mechanisms that regulate hOPC homeostasis remain poorly defined. Herein, we identify paired related homeobox protein 1 (PRRX1) in primary PDGFαR+ hOPCs. We show that enforced PRRX1 expression results in reversible G1/0 arrest. While both PRRX1 splice variants reduce hOPC proliferation, only PRRX1a abrogates migration. hOPC engraftment into hypomyelinated shiverer/rag2 mouse brain is severely impaired by PRRX1a, characterized by reduced cell proliferation and migration. PRRX1 induces a gene expression signature characteristic of stem cell quiescence. Both IFN-γ and BMP signaling upregulate PRRX1 and induce quiescence. PRRX1 knockdown modulates IFN-γ-induced quiescence. In mouse brain, PRRX1 mRNA was detected in non-dividing OPCs and is upregulated in OPCs following demyelination. Together, these data identify PRRX1 as a regulator of quiescence in hOPCs and as a potential regulator of pathological quiescence.


Asunto(s)
Ciclo Celular , Proteínas de Homeodominio/metabolismo , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Animales , Proteínas Morfogenéticas Óseas/farmacología , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interferón gamma/farmacología , Antígeno Ki-67/metabolismo , Ratones , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/trasplante , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
3.
J Neurosci ; 38(31): 6921-6932, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29959237

RESUMEN

Muscarinic receptor antagonists act as potent inducers of oligodendrocyte differentiation and accelerate remyelination. However, the use of muscarinic antagonists in the clinic is limited by poor understanding of the operant receptor subtype, and questions regarding possible species differences between rodents and humans. Based on high selective expression in human oligodendrocyte progenitor cells (OPCs), we hypothesized that M3R is the functionally relevant receptor. Lentiviral M3R knockdown in human primary CD140a/PDGFαR+ OPCs resulted in enhanced differentiation in vitro and substantially reduced the calcium response following muscarinic agonist treatment. Importantly, following transplantation in hypomyelinating shiverer/rag2 mice, M3R knockdown improved remyelination by human OPCs. Furthermore, conditional M3R ablation in adult NG2-expressing OPCs increased oligodendrocyte differentiation and led to improved spontaneous remyelination in mice. Together, we demonstrate that M3R receptor mediates muscarinic signaling in human OPCs that act to delay differentiation and remyelination, suggesting that M3 receptors are viable targets for human demyelinating disease.SIGNIFICANCE STATEMENT The identification of drug targets aimed at improving remyelination in patients with demyelination disease is a key step in development of effective regenerative therapies to treat diseases, such as multiple sclerosis. Muscarinic receptor antagonists have been identified as effective potentiators of remyelination, but the receptor subtypes that mediate these receptors are unclear. In this study, we show that genetic M3R ablation in both mouse and human cells results in improved remyelination and is mediated by acceleration of oligodendrocyte commitment from oligodendrocyte progenitor cells. Therefore, M3R represents an attractive target for induced remyelination in human disease.


Asunto(s)
Vaina de Mielina/fisiología , Neurogénesis/fisiología , Células Precursoras de Oligodendrocitos/fisiología , Receptor Muscarínico M3/fisiología , Remielinización/fisiología , Animales , Trasplante de Tejido Encefálico , Señalización del Calcio , Células Cultivadas , Trasplante de Tejido Fetal , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Mutantes Neurológicos , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Prosencéfalo/embriología , Prosencéfalo/trasplante , Interferencia de ARN , Receptor Muscarínico M3/agonistas , Receptor Muscarínico M3/antagonistas & inhibidores , Médula Espinal/química , Médula Espinal/ultraestructura
4.
Stem Cell Reports ; 9(2): 710-723, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28793249

RESUMEN

Impaired human oligodendrocyte progenitor cell (hOPC) differentiation likely contributes to failed remyelination in multiple sclerosis. The characterization of molecular pathways that regulate hOPC differentiation will provide means to induce remyelination. In this study, we determined the gene expression profile of PDGFαR+ hOPCs during initial oligodendrocyte commitment. Weighted gene coexpression network analysis was used to define progenitor and differentiation-specific gene expression modules and functionally important hub genes. These modules were compared with rodent OPC and oligodendrocyte data to determine the extent of species conservation. These analyses identified G-protein ß4 (GNB4), which was associated with hOPC commitment. Lentiviral GNB4 overexpression rapidly induced human oligodendrocyte differentiation. Following xenograft in hypomyelinating shiverer/rag2 mice, GNB4 overexpression augmented myelin synthesis and the ability of hOPCs to ensheath host axons, establishing GNB4 as functionally important in human myelination. As such, network analysis of hOPC gene expression accurately predicts genes that influence human oligodendrocyte differentiation in vivo.


Asunto(s)
Diferenciación Celular/genética , Biología Computacional/métodos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genómica , Células Precursoras de Oligodendrocitos/citología , Células Precursoras de Oligodendrocitos/metabolismo , Animales , Axones/metabolismo , Subunidades beta de la Proteína de Unión al GTP/genética , Perfilación de la Expresión Génica , Genómica/métodos , Humanos , Oligodendroglía/citología , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Roedores , Transducción de Señal , Transcriptoma
5.
J Neurosci ; 35(8): 3676-88, 2015 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-25716865

RESUMEN

Therapeutic repair of myelin disorders may be limited by the relatively slow rate of human oligodendrocyte differentiation. To identify appropriate pharmacological targets with which to accelerate differentiation of human oligodendrocyte progenitors (hOPCs) directly, we used CD140a/O4-based FACS of human forebrain and microarray to hOPC-specific receptors. Among these, we identified CHRM3, a M3R muscarinic acetylcholine receptor, as being restricted to oligodendrocyte-biased CD140a(+)O4(+) cells. Muscarinic agonist treatment of hOPCs resulted in a specific and dose-dependent blockade of oligodendrocyte commitment. Conversely, when hOPCs were cocultured with human neurons, M3R antagonist treatment stimulated oligodendrocytic differentiation. Systemic treatment with solifenacin, an FDA-approved muscarinic receptor antagonist, increased oligodendrocyte differentiation of transplanted hOPCs in hypomyelinated shiverer/rag2 brain. Importantly, solifenacin treatment of engrafted animals reduced auditory brainstem response interpeak latency, indicative of increased conduction velocity and thereby enhanced functional repair. Therefore, solifenacin and other selective muscarinic antagonists represent new adjunct approaches to accelerate repair by engrafted human progenitors.


Asunto(s)
Células Madre Fetales/citología , Antagonistas Muscarínicos/farmacología , Vaina de Mielina/metabolismo , Oligodendroglía/citología , Quinuclidinas/farmacología , Regeneración , Tetrahidroisoquinolinas/farmacología , Animales , Tronco Encefálico/citología , Tronco Encefálico/fisiología , Células Cultivadas , Proteínas de Unión al ADN/genética , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Células Madre Fetales/efectos de los fármacos , Células Madre Fetales/metabolismo , Células Madre Fetales/trasplante , Humanos , Masculino , Ratones , Agonistas Muscarínicos/farmacología , Vaina de Mielina/genética , Neurogénesis , Antígenos O/genética , Antígenos O/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/trasplante , Prosencéfalo/citología , Prosencéfalo/embriología , Receptor Muscarínico M3 , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Succinato de Solifenacina
6.
Stem Cell Reports ; 3(2): 250-9, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25254339

RESUMEN

Multiple sclerosis (MS) is a chronic demyelinating disease of unknown etiology that affects the CNS. While current therapies are primarily directed against the immune system, the new challenge is to address progressive MS with remyelinating and neuroprotective strategies. Here, we develop a highly reproducible protocol to efficiently derive oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes from induced pluripotent stem cells (iPSCs). Key elements of our protocol include adherent cultures, dual SMAD inhibition, and addition of retinoids from the beginning of differentiation, which lead to increased yields of OLIG2 progenitors and high numbers of OPCs within 75 days. Furthermore, we show the generation of viral and integration-free iPSCs from primary progressive MS (PPMS) patients and their efficient differentiation to oligodendrocytes. PPMS OPCs are functional, as demonstrated by in vivo myelination in the shiverer mouse. These results provide encouraging advances toward the development of autologous cell therapies using iPSCs.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Esclerosis Múltiple/patología , Oligodendroglía/citología , Animales , Axones/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Oligodendroglía/trasplante , Proteínas Smad/antagonistas & inhibidores , Proteínas Smad/metabolismo , Trasplante Heterólogo , Tretinoina/farmacología
7.
Proc Natl Acad Sci U S A ; 111(28): E2885-94, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24982138

RESUMEN

Human oligodendrocyte progenitor cell (OPC) specification and differentiation occurs slowly and limits the potential for cell-based treatment of demyelinating disease. In this study, using FACS-based isolation and microarray analysis, we identified a set of transcription factors expressed by human primary CD140a(+)O4(+) OPCs relative to CD133(+)CD140a(-) neural stem/progenitor cells (NPCs). Among these, lentiviral overexpression of transcription factors ASCL1, SOX10, and NKX2.2 in NPCs was sufficient to induce Sox10 enhancer activity, OPC mRNA, and protein expression consistent with OPC fate; however, unlike ASCL1 and NKX2.2, only the transcriptome of SOX10-infected NPCs was induced to a human OPC gene expression signature. Furthermore, only SOX10 promoted oligodendrocyte commitment, and did so at quantitatively equivalent levels to native OPCs. In xenografts of shiverer/rag2 animals, SOX10 increased the rate of mature oligodendrocyte differentiation and axon ensheathment. Thus, SOX10 appears to be the principle and rate-limiting regulator of myelinogenic fate from human NPCs.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Elementos de Facilitación Genéticos , Xenoinjertos , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio , Humanos , Lentivirus , Ratones , Células-Madre Neurales/citología , Proteínas Nucleares , Oligodendroglía/citología , Trasplante de Células Madre , Factores de Transcripción/genética , Transcriptoma , Transducción Genética
8.
Exp Neurol ; 247: 694-702, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23507034

RESUMEN

In this study, we sought to establish a novel method to prospectively and dynamically identify live human oligodendrocyte precursor cells (OPCs) and oligodendrocyte lineage cells from brain dissociates and pluripotent stem cell culture. We selected a highly conserved enhancer element of the Sox10 gene, known as MCS5, which directs reporter expression to oligodendrocyte lineage cells in mouse and zebrafish. We demonstrate that lentiviral Sox10-MCS5 induced expression of GFP at high levels in a subpopulation of human CD140a/PDGFαR-sorted OPCs as well as their immature oligodendrocyte progeny. Furthermore, we show that almost all Sox10-MCS5:GFP(high) cells expressed OPC antigen CD140a and human OPCs expressing SOX10, OLIG2, and PDGFRA mRNAs could be prospectively identified using GFP based fluorescence activated cells sorting alone. Additionally, we established a human induced pluripotent cell (iPSC) line transduced with the Sox10-MCS5:GFP reporter using a Rex-Neo cassette. Similar to human primary cells, GFP expression was restricted to embryoid bodies containing both oligodendrocyte progenitor and oligodendrocyte cells and co-localized with NG2 and O4-positive cells respectively. As such, we have developed a novel reporter system that can track oligodendrocyte commitment in human cells, establishing a valuable tool to improve our understanding and efficiency of human oligodendrocyte derivation.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/fisiología , Elementos de Facilitación Genéticos/genética , Oligodendroglía/metabolismo , Factores de Transcripción SOXE/metabolismo , Antígenos/metabolismo , Células Cultivadas , Feto , Citometría de Flujo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Antígenos O/metabolismo , Proteoglicanos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción SOXE/genética
9.
Stem Cells Dev ; 22(15): 2121-31, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23488628

RESUMEN

The mechanisms underlying the specification of oligodendrocyte fate from multipotent neural progenitor cells (NPCs) in developing human brain are unknown. In this study, we sought to identify antigens sufficient to distinguish NPCs free from oligodendrocyte progenitor cells (OPCs). We investigated the potential overlap of NPC and OPC antigens using multicolor fluorescence-activated cell sorting (FACS) for CD133/PROM1, A2B5, and CD140a/PDGFαR antigens. Surprisingly, we found that CD133, but not A2B5, was capable of enriching for OLIG2 expression, Sox10 enhancer activity, and oligodendrocyte potential. As a subpopulation of CD133-positive cells expressed CD140a, we asked whether CD133 enriched bone fide NPCs regardless of CD140a expression. We found that CD133(+)CD140a(-) cells were highly enriched for neurosphere initiating cells and were multipotent. Importantly, when analyzed immediately following isolation, CD133(+)CD140a(-) NPCs lacked the capacity to generate oligodendrocytes. In contrast, CD133(+)CD140a(+) cells were OLIG2-expressing OPCs capable of oligodendrocyte differentiation, but formed neurospheres with lower efficiency and were largely restricted to glial fate. Gene expression analysis further confirmed the stem cell nature of CD133(+)CD140a(-) cells. As human CD133(+) cells comprised both NPCs and OPCs, CD133 expression alone cannot be considered a specific marker of the stem cell phenotype, but rather comprises a heterogeneous mix of glial restricted as well as multipotent neural precursors. In contrast, CD133/CD140a-based FACS permits the separation of defined progenitor populations and the study of neural stem and oligodendrocyte fate specification in the human brain.


Asunto(s)
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Péptidos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Antígeno AC133 , Biomarcadores/metabolismo , Separación Celular , Células Cultivadas , Citometría de Flujo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Transcriptoma
10.
Glia ; 60(12): 1944-53, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22927334

RESUMEN

The molecular mechanisms controlling human oligodendrocyte development are poorly characterized. Microarray analysis of human oligodendrocyte progenitor cells (OPCs) and immature oligodendrocytes revealed that specific-class I histone deacetylase (HDAC) target genes were actively repressed during oligodendrocyte commitment. Although epigenetic regulation of oligodendrocyte differentiation has been established in rodent development, the role of HDACs in human OPCs remains undefined. We used HDAC inhibitors (HDACi) trichostatin A (TSA) and sodium butyrate to determine the importance of HDAC activity in human primary OPC differentiation. Treatment with either drug resulted in significant dose-dependent inhibition of O4(+) oligodendrocyte cell differentiation, reduction of oligodendrocyte morphological maturation, and downregulation of myelin basic protein mRNA. High dose TSA treatment was also associated with reduction in OPC proliferation. HDACi treatment prevented downregulation of SOX2, ID4, and TCF7L2 mRNAs but did not regulate HES5, suggesting that targets of HDAC repression may differ between species. These results predict that HDACi treatment would impair proliferation and differentiation by parenchymal oligodendrocyte progenitors, and thereby degrade their potential for endogenous repair in human demyelinating disease. © 2012 Wiley Periodicals, Inc.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/enzimología , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Oligodendroglía/enzimología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Feto/citología , Feto/efectos de los fármacos , Feto/enzimología , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/fisiología , Humanos , Oligodendroglía/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Células Madre/efectos de los fármacos , Células Madre/enzimología
11.
Neurotoxicol Teratol ; 28(5): 617-24, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16989981

RESUMEN

BACKGROUND: Valproic acid (VPA) exposure in utero has been associated with an increased risk of both neural tube defects and autism spectrum disorders (ASDs). The terata induced by VPA suggest interference with pattern formation. Retinoic acid produces similar terata and is known to act in part by increasing the expression of Hoxa1. We tested the hypotheses that exposure to VPA would alter the expression of Hoxa1 in rat embryos during times of normal Hoxa1 expression (d10.5-13.5) and that exposure at earlier and later stages would induce inappropriate expression. METHOD: Hoxa1 expression levels were determined by real-time PCR in individual embryos 1 h after exposure on gestational d10, 12, 13, 14, or 15. Additionally, teratogenic (4-yn-VPA) and nonteratogenic analogs of VPA (IE-VPA), retinoic acid (RA), and saline were compared for effects on Hoxa1 expression on d12. Embryos were allowed to develop for 1, 2, 4, 6, or 24 h, to follow the time course of effects. RESULTS: In utero exposure to VPA on gestational d10 and on d12-14 significantly increased the level of Hoxa1 expression compared to saline-exposed embryos at developmental ages prior to, during and after the normal expression period for this gene. On gestational d12, exposures to VPA and 4-yn-VPA significantly increased Hoxa1 expression at all sacrifice times, compared to saline-exposed embryos. RA significantly elevated Hoxa1 expression at all time points except 24-h post-treatment. The nonteratogenic VPA analog, IE-VPA, did not affect Hoxa1 expression. CONCLUSIONS: VPA and 4-yn-VPA exposures elevated Hoxa1 mRNA during its normal expression period and induced expression outside of the normal period. This may explain, in part, how VPA disrupts development.


Asunto(s)
Inhibidores Enzimáticos/toxicidad , Ácidos Grasos Insaturados/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Efectos Tardíos de la Exposición Prenatal , Ácido Valproico/toxicidad , Animales , Ácidos Grasos Insaturados/química , Femenino , Masculino , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Long-Evans , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Factores de Tiempo
12.
Biol Psychiatry ; 55(4): 413-9, 2004 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-14960295

RESUMEN

BACKGROUND: The HOXA1 gene plays a major role in brainstem and cranial morphogenesis. The G allele of the HOXA1 A218G polymorphism has been previously found associated with autism. METHODS: We performed case-control and family-based association analyses, contrasting 127 autistic patients with 174 ethnically matched controls, and assessing for allelic transmission disequilibrium in 189 complete trios. RESULTS: A, and not G, alleles were associated with autism using both case-control (chi(2) = 8.96 and 5.71, 1 df, p <.005 and <.025 for genotypes and alleles, respectively), and family-based (transmission/disequilibrium test chi(2) = 8.80, 1 df, p <.005) association analyses. The head circumference of 31 patients carrying one or two copies of the G allele displayed significantly larger median values (95.0th vs. 82.5th percentile, p <.05) and dramatically reduced interindividual variability (p <.0001), compared with 166 patients carrying the A/A genotype. CONCLUSIONS: The HOXA1 A218G polymorphism explains approximately 5% of the variance in the head circumference of autistic patients and represents to our knowledge the first known gene variant providing sizable contributions to cranial morphology. The disease specificity of this finding is currently being investigated. Nonreplications in genetic linkage/association studies could partly stem from the dyshomogeneous distribution of an endophenotype morphologically defined by cranial circumference.


Asunto(s)
Trastorno Autístico/genética , Proteínas de Homeodominio/genética , Polimorfismo Genético , Factores de Transcripción/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Alanina/genética , Américas , Síndrome de Asperger , Trastorno Autístico/patología , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Niño , Preescolar , Análisis Mutacional de ADN , Salud de la Familia , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Glicina/genética , Cabeza/patología , Humanos , Italia , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Base del Cráneo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...