Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Science ; 381(6658): eade6289, 2023 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-37561850

RESUMEN

Skin color, one of the most diverse human traits, is determined by the quantity, type, and distribution of melanin. In this study, we leveraged the light-scattering properties of melanin to conduct a genome-wide screen for regulators of melanogenesis. We identified 169 functionally diverse genes that converge on melanosome biogenesis, endosomal transport, and gene regulation, of which 135 represented previously unknown associations with pigmentation. In agreement with their melanin-promoting function, the majority of screen hits were up-regulated in melanocytes from darkly pigmented individuals. We further unraveled functions of KLF6 as a transcription factor that regulates melanosome maturation and pigmentation in vivo, and of the endosomal trafficking protein COMMD3 in modulating melanosomal pH. Our study reveals a plethora of melanin-promoting genes, with broad implications for human variation, cell biology, and medicine.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Factor 6 Similar a Kruppel , Melaninas , Melanocitos , Melanosomas , Pigmentación de la Piel , Humanos , Melaninas/biosíntesis , Melaninas/genética , Melanocitos/metabolismo , Melanosomas/metabolismo , Pigmentación de la Piel/genética , Estudio de Asociación del Genoma Completo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor 6 Similar a Kruppel/genética , Factor 6 Similar a Kruppel/metabolismo , Endosomas/metabolismo , Animales , Ratones , Línea Celular Tumoral
2.
Sci Adv ; 6(12): eaaz9115, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32219172

RESUMEN

Zinc finger protein Zscan4 is selectively expressed in mouse two-cell (2C) embryos undergoing zygotic genome activation (ZGA) and in a rare subpopulation of embryonic stem cells with 2C-like features. Here, we show that Zscan4 specifically recognizes a subset of (CA)n microsatellites, repeat sequences prone to genomic instability. Zscan4-associated microsatellite regions are characterized by low nuclease sensitivity and high histone occupancy. In vitro, Zscan4 binds nucleosomes and protects them from disassembly upon torsional strain. Furthermore, Zscan4 depletion leads to elevated DNA damage in 2C mouse embryos in a transcription-dependent manner. Together, our results identify Zscan4 as a DNA sequence-dependent microsatellite binding factor and suggest a developmentally regulated mechanism, which protects fragile genomic regions from DNA damage at a time of embryogenesis associated with high transcriptional burden and genomic stress.


Asunto(s)
Daño del ADN , Células Madre Embrionarias/metabolismo , Repeticiones de Microsatélite , Factores de Transcripción/metabolismo , Dedos de Zinc , Animales , Sitios de Unión , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Ratones , Modelos Biológicos , Nucleosomas/metabolismo , Motivos de Nucleótidos , Unión Proteica , Secuencias Repetitivas de Ácidos Nucleicos
3.
Cell ; 178(6): 1421-1436.e24, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31491386

RESUMEN

The developmental disorder Floating-Harbor syndrome (FHS) is caused by heterozygous truncating mutations in SRCAP, a gene encoding a chromatin remodeler mediating incorporation of histone variant H2A.Z. Here, we demonstrate that FHS-associated mutations result in loss of SRCAP nuclear localization, alter neural crest gene programs in human in vitro models and Xenopus embryos, and cause craniofacial defects. These defects are mediated by one of two H2A.Z subtypes, H2A.Z.2, whose knockdown mimics and whose overexpression rescues the FHS phenotype. Selective rescue by H2A.Z.2 is conferred by one of the three amino acid differences between the H2A.Z subtypes, S38/T38. We further show that H2A.Z.1 and H2A.Z.2 genomic occupancy patterns are qualitatively similar, but quantitatively distinct, and H2A.Z.2 incorporation at AT-rich enhancers and expression of their associated genes are both sensitized to SRCAP truncations. Altogether, our results illuminate the mechanism underlying a human syndrome and uncover selective functions of H2A.Z subtypes during development.


Asunto(s)
Anomalías Múltiples/genética , Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Anomalías Craneofaciales/genética , Trastornos del Crecimiento/genética , Defectos del Tabique Interventricular/genética , Histonas/genética , Adenosina Trifosfatasas/genética , Sustitución de Aminoácidos , Animales , Células Madre Embrionarias , Células HEK293 , Humanos , Mutación , Xenopus laevis
4.
Elife ; 72018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-30070637

RESUMEN

Recent work suggests extensive adaptation of transposable elements (TEs) for host gene regulation. However, high numbers of integrations typical of TEs, coupled with sequence divergence within families, have made systematic interrogation of the regulatory contributions of TEs challenging. Here, we employ CARGO, our recent method for CRISPR gRNA multiplexing, to facilitate targeting of LTR5HS, an ape-specific class of HERVK (HML-2) LTRs that is active during early development and present in ~700 copies throughout the human genome. We combine CARGO with CRISPR activation or interference to, respectively, induce or silence LTR5HS en masse, and demonstrate that this system robustly targets the vast majority of LTR5HS insertions. Remarkably, activation/silencing of LTR5HS is associated with reciprocal up- and down-regulation of hundreds of human genes. These effects require the presence of retroviral sequences, but occur over long genomic distances, consistent with a pervasive function of LTR5HS elements as early embryonic enhancers in apes.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica/genética , Genoma Humano/genética , Secuencias Repetidas Terminales/genética , Animales , Elementos Transponibles de ADN , Retrovirus Endógenos/genética , Hominidae/genética , Humanos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retroviridae/genética
5.
Nature ; 554(7690): 112-117, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364875

RESUMEN

Many craniofacial disorders are caused by heterozygous mutations in general regulators of housekeeping cellular functions such as transcription or ribosome biogenesis. Although it is understood that many of these malformations are a consequence of defects in cranial neural crest cells, a cell type that gives rise to most of the facial structures during embryogenesis, the mechanism underlying cell-type selectivity of these defects remains largely unknown. By exploring molecular functions of DDX21, a DEAD-box RNA helicase involved in control of both RNA polymerase (Pol) I- and II-dependent transcriptional arms of ribosome biogenesis, we uncovered a previously unappreciated mechanism linking nucleolar dysfunction, ribosomal DNA (rDNA) damage, and craniofacial malformations. Here we demonstrate that genetic perturbations associated with Treacher Collins syndrome, a craniofacial disorder caused by heterozygous mutations in components of the Pol I transcriptional machinery or its cofactor TCOF1 (ref. 1), lead to relocalization of DDX21 from the nucleolus to the nucleoplasm, its loss from the chromatin targets, as well as inhibition of rRNA processing and downregulation of ribosomal protein gene transcription. These effects are cell-type-selective, cell-autonomous, and involve activation of p53 tumour-suppressor protein. We further show that cranial neural crest cells are sensitized to p53-mediated apoptosis, but blocking DDX21 loss from the nucleolus and chromatin rescues both the susceptibility to apoptosis and the craniofacial phenotypes associated with Treacher Collins syndrome. This mechanism is not restricted to cranial neural crest cells, as blood formation is also hypersensitive to loss of DDX21 functions. Accordingly, ribosomal gene perturbations associated with Diamond-Blackfan anaemia disrupt DDX21 localization. At the molecular level, we demonstrate that impaired rRNA synthesis elicits a DNA damage response, and that rDNA damage results in tissue-selective and dosage-dependent effects on craniofacial development. Taken together, our findings illustrate how disruption in general regulators that compromise nucleolar homeostasis can result in tissue-selective malformations.


Asunto(s)
Nucléolo Celular/metabolismo , Nucléolo Celular/patología , Daño del ADN , ADN Ribosómico/metabolismo , Disostosis Mandibulofacial/genética , Disostosis Mandibulofacial/patología , Estrés Fisiológico , Animales , Apoptosis , Benzotiazoles/farmacología , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patología , Cromatina/metabolismo , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , ADN Ribosómico/genética , ARN Polimerasas Dirigidas por ADN/deficiencia , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Disostosis Mandibulofacial/embriología , Ratones , Naftiridinas/farmacología , Cresta Neural/enzimología , Cresta Neural/patología , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Fenotipo , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , ARN Helicasas/metabolismo , ARN Polimerasa I/antagonistas & inhibidores , ARN Ribosómico/biosíntesis , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Proteínas Ribosómicas/biosíntesis , Proteínas Ribosómicas/genética , Ribosomas/genética , Ribosomas/metabolismo , Cráneo/patología , Estrés Fisiológico/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Xenopus , Pez Cebra/embriología , Proteínas de Pez Cebra/deficiencia
6.
Cell ; 170(4): 774-786.e19, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28802045

RESUMEN

Conflicts between transcription and replication are a potent source of DNA damage. Co-transcriptional R-loops could aggravate such conflicts by creating an additional barrier to replication fork progression. Here, we use a defined episomal system to investigate how conflict orientation and R-loop formation influence genome stability in human cells. R-loops, but not normal transcription complexes, induce DNA breaks and orientation-specific DNA damage responses during conflicts with replication forks. Unexpectedly, the replisome acts as an orientation-dependent regulator of R-loop levels, reducing R-loops in the co-directional (CD) orientation but promoting their formation in the head-on (HO) orientation. Replication stress and deregulated origin firing increase the number of HO collisions leading to genome-destabilizing R-loops. Our findings connect DNA replication to R-loop homeostasis and suggest a mechanistic basis for genome instability resulting from deregulated DNA replication, observed in cancer and other disease states.


Asunto(s)
Replicación del ADN , Transcripción Genética , Daño del ADN , Momento de Replicación del ADN , Inestabilidad Genómica , Células HEK293 , Humanos , Plásmidos
7.
Cancer Cell ; 31(5): 635-652.e6, 2017 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-28434841

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric cancer with limited therapeutic options. The majority of cases of DIPG exhibit a mutation in histone-3 (H3K27M) that results in oncogenic transcriptional aberrancies. We show here that DIPG is vulnerable to transcriptional disruption using bromodomain inhibition or CDK7 blockade. Targeting oncogenic transcription through either of these methods synergizes with HDAC inhibition, and DIPG cells resistant to HDAC inhibitor therapy retain sensitivity to CDK7 blockade. Identification of super-enhancers in DIPG provides insights toward the cell of origin, highlighting oligodendroglial lineage genes, and reveals unexpected mechanisms mediating tumor viability and invasion, including potassium channel function and EPH receptor signaling. The findings presented demonstrate transcriptional vulnerabilities and elucidate previously unknown mechanisms of DIPG pathobiology.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Azepinas/farmacología , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Fenilendiaminas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Transcripción Genética/efectos de los fármacos , Triazoles/farmacología , Animales , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/metabolismo , Neoplasias del Tronco Encefálico/patología , Proteínas de Ciclo Celular , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Glioma/genética , Glioma/metabolismo , Glioma/patología , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Mutación , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Panobinostat , Cultivo Primario de Células , Interferencia de ARN , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Activadora de Quinasas Ciclina-Dependientes
8.
Nat Commun ; 8: 14802, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28378740

RESUMEN

CD47 is a cell surface molecule that inhibits phagocytosis of cells that express it by binding to its receptor, SIRPα, on macrophages and other immune cells. CD47 is expressed at different levels by neoplastic and normal cells. Here, to reveal mechanisms by which different neoplastic cells generate this dominant 'don't eat me' signal, we analyse the CD47 regulatory genomic landscape. We identify two distinct super-enhancers (SEs) associated with CD47 in certain cancer cell types. We show that a set of active constituent enhancers, located within the two CD47 SEs, regulate CD47 expression in different cancer cell types and that disruption of CD47 SEs reduces CD47 gene expression. Finally we report that the TNF-NFKB1 signalling pathway directly regulates CD47 by interacting with a constituent enhancer located within a CD47-associated SE specific to breast cancer. These results suggest that cancers can evolve SE to drive CD47 overexpression to escape immune surveillance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antígeno CD47/fisiología , Elementos de Facilitación Genéticos , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Transducción de Señal , Regulación hacia Arriba , Animales , Neoplasias de la Mama/patología , Antígeno CD47/genética , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Subunidad p50 de NF-kappa B/metabolismo , Fagocitosis , Unión Proteica , Factor de Necrosis Tumoral alfa/metabolismo
9.
Elife ; 52016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27552054

RESUMEN

The hormone estrogen (E2) binds the estrogen receptor to promote transcription of E2-responsive genes in the breast and other tissues. E2 also has links to genomic instability, and elevated E2 levels are tied to breast cancer. Here, we show that E2 stimulation causes a rapid, global increase in the formation of R-loops, co-transcriptional RNA-DNA products, which in some instances have been linked to DNA damage. We show that E2-dependent R-loop formation and breast cancer rearrangements are highly enriched at E2-responsive genomic loci and that E2 induces DNA replication-dependent double-strand breaks (DSBs). Strikingly, many DSBs that accumulate in response to E2 are R-loop dependent. Thus, R-loops resulting from the E2 transcriptional response are a significant source of DNA damage. This work reveals a novel mechanism by which E2 stimulation leads to genomic instability and highlights how transcriptional programs play an important role in shaping the genomic landscape of DNA damage susceptibility.


Asunto(s)
Daño del ADN , Estrógenos/toxicidad , Mutágenos/metabolismo , Transcripción Genética , ADN/metabolismo , Roturas del ADN de Doble Cadena , Humanos , Células MCF-7 , ARN Mensajero/metabolismo
10.
Nature ; 458(7237): 529-33, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19212323

RESUMEN

Epigenetic mechanisms that maintain neurogenesis throughout adult life remain poorly understood. Trithorax group (trxG) and Polycomb group (PcG) gene products are part of an evolutionarily conserved chromatin remodelling system that activate or silence gene expression, respectively. Although PcG member Bmi1 has been shown to be required for postnatal neural stem cell self-renewal, the role of trxG genes remains unknown. Here we show that the trxG member Mll1 (mixed-lineage leukaemia 1) is required for neurogenesis in the mouse postnatal brain. Mll1-deficient subventricular zone neural stem cells survive, proliferate and efficiently differentiate into glial lineages; however, neuronal differentiation is severely impaired. In Mll1-deficient cells, early proneural Mash1 (also known as Ascl1) and gliogenic Olig2 expression are preserved, but Dlx2, a key downstream regulator of subventricular zone neurogenesis, is not expressed. Overexpression of Dlx2 can rescue neurogenesis in Mll1-deficient cells. Chromatin immunoprecipitation demonstrates that Dlx2 is a direct target of MLL in subventricular zone cells. In differentiating wild-type subventricular zone cells, Mash1, Olig2 and Dlx2 loci have high levels of histone 3 trimethylated at lysine 4 (H3K4me3), consistent with their transcription. In contrast, in Mll1-deficient subventricular zone cells, chromatin at Dlx2 is bivalently marked by both H3K4me3 and histone 3 trimethylated at lysine 27 (H3K27me3), and the Dlx2 gene fails to properly activate. These data support a model in which Mll1 is required to resolve key silenced bivalent loci in postnatal neural precursors to the actively transcribed state for the induction of neurogenesis, but not for gliogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neurogénesis , Neuronas/citología , Células Madre/citología , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Metilación , Ratones , Proteína de la Leucemia Mieloide-Linfoide/deficiencia , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/metabolismo , Bulbo Olfatorio/citología , Bulbo Olfatorio/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Células Madre/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Cell ; 139(7): 1290-302, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064375

RESUMEN

Polycomb Repressive Complex 2 (PRC2) regulates key developmental genes in embryonic stem (ES) cells and during development. Here we show that Jarid2/Jumonji, a protein enriched in pluripotent cells and a founding member of the Jumonji C (JmjC) domain protein family, is a PRC2 subunit in ES cells. Genome-wide ChIP-seq analyses of Jarid2, Ezh2, and Suz12 binding reveal that Jarid2 and PRC2 occupy the same genomic regions. We further show that Jarid2 promotes PRC2 recruitment to the target genes while inhibiting PRC2 histone methyltransferase activity, suggesting that it acts as a "molecular rheostat" that finely calibrates PRC2 functions at developmental genes. Using Xenopus laevis as a model we demonstrate that Jarid2 knockdown impairs the induction of gastrulation genes in blastula embryos and results in failure of differentiation. Our findings illuminate a mechanism of histone methylation regulation in pluripotent cells and during early cell-fate transitions.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/metabolismo , Animales , Células Madre Embrionarias/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mitocondrias/metabolismo , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , ARN/metabolismo , Proteína 2 de Unión a Retinoblastoma/metabolismo
12.
Nature ; 442(7098): 86-90, 2006 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-16728976

RESUMEN

Lysine methylation of histones is recognized as an important component of an epigenetic indexing system demarcating transcriptionally active and inactive chromatin domains. Trimethylation of histone H3 lysine 4 (H3K4me3) marks transcription start sites of virtually all active genes. Recently, we reported that the WD40-repeat protein WDR5 is important for global levels of H3K4me3 and control of HOX gene expression. Here we show that a plant homeodomain (PHD) finger of nucleosome remodelling factor (NURF), an ISWI-containing ATP-dependent chromatin-remodelling complex, mediates a direct preferential association with H3K4me3 tails. Depletion of H3K4me3 causes partial release of the NURF subunit, BPTF (bromodomain and PHD finger transcription factor), from chromatin and defective recruitment of the associated ATPase, SNF2L (also known as ISWI and SMARCA1), to the HOXC8 promoter. Loss of BPTF in Xenopus embryos mimics WDR5 loss-of-function phenotypes, and compromises spatial control of Hox gene expression. These results strongly suggest that WDR5 and NURF function in a common biological pathway in vivo, and that NURF-mediated ATP-dependent chromatin remodelling is directly coupled to H3K4 trimethylation to maintain Hox gene expression patterns during development. We also identify a previously unknown function for the PHD finger as a highly specialized methyl-lysine-binding domain.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Adenosina Trifosfatasas/metabolismo , Secuencias de Aminoácidos , Animales , Antígenos Nucleares , Epigénesis Genética , Regulación de la Expresión Génica , Humanos , Metilación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Xenopus/embriología , Xenopus/crecimiento & desarrollo , Xenopus/metabolismo
13.
J Virol ; 79(16): 10650-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16051857

RESUMEN

Nef proteins of primate lentiviruses promote viral replication, virion infectivity, and evasion of antiviral immune responses by modulating signal transduction pathways and downregulating expression of receptors at the cell surface that are important for efficient antigen-specific responses, such as CD4, CD28, T-cell antigen receptor, and class I and class II major histocompatibility complex. Here we show that Nef proteins from diverse groups of primate lentiviruses which do not require the chemokine receptor CXCR4 for entry into target cells strongly downmodulate the cell surface expression of CXCR4. In contrast, all human immunodeficiency virus type 1 (HIV-1) and the majority of HIV-2 Nef proteins tested did not have such strong effects. SIVmac239 Nef strongly inhibited lymphocyte migration to CXCR4 ligand, the chemokine stromal derived factor 1 (SDF-1). SIVmac239 Nef downregulated CXCR4 by accelerating the rate of its endocytosis. Downmodulation of CXCR4 was abolished by mutations that disrupt the constitutively strong AP-2 clathrin adaptor binding element located in the N-terminal region of the Nef molecule, suggesting that Nef accelerates CXCR4 endocytosis via an AP-2-dependent pathway. Together, these results point to CXCR4 as playing an important role in simian immunodeficiency virus and possibly also HIV-2 persistence in vivo that is unrelated to viral entry into target cells. We speculate that Nef targets CXCR4 to disrupt ordered trafficking of infected leukocytes between local microenvironments in order to facilitate their dissemination and/or impair the antiviral immune response.


Asunto(s)
Quimiocinas CXC/fisiología , Productos del Gen nef/fisiología , VIH/fisiología , Receptores CXCR4/antagonistas & inhibidores , Virus de la Inmunodeficiencia de los Simios/fisiología , Antígenos CD4/fisiología , Movimiento Celular , Quimiocina CXCL12 , Regulación hacia Abajo , Endocitosis , Productos del Gen nef/química , Humanos , Células Jurkat , Receptores CXCR4/fisiología , Linfocitos T/fisiología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana
14.
Blood ; 102(8): 2925-32, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12855553

RESUMEN

The HIV-1 gene nef is important for progression toward AIDS and cellular depletion of the infected thymus. Expression of the Nef protein alone impairs human thymopoiesis. Here, we performed a structure-function analysis of the Nef protein by comparing the effect on T-cell development of different nef alleles, either wild type or defective for selected functions, expressed by human thymocytes. We show that Nef-mediated impaired thymopoiesis is not due to altered surface marker trafficking, nor dependent on oligomerization of Nef. By contrast, mutations in the myristoylation site and in signaling sites of Nef, ie, sites important for interaction with phosphofurin acidic cluster sorting protein-1 (PACS-1), Src homology domain 3 (SH3) domains, and p21-activated kinase 2 (PAK2), were found to be critical for its effect on T-cell development. These results point to sites in Nef to target therapeutically for restoration of thymopoiesis in HIV infection.


Asunto(s)
Productos del Gen nef/metabolismo , Linfocitos T/metabolismo , Timo/citología , Alelos , Anticuerpos Monoclonales/metabolismo , Antígenos CD34/biosíntesis , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Separación Celular , Dimerización , Regulación hacia Abajo , Citometría de Flujo , Técnicas de Transferencia de Gen , Humanos , Mutación , Ácido Mirístico/metabolismo , Plásmidos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Retroviridae/genética , Retroviridae/metabolismo , Transducción de Señal , Células Madre/metabolismo , Timo/metabolismo , Proteínas de Transporte Vesicular , Quinasas p21 Activadas , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA