Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.307
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 176(1-2): 167-181.e21, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30595447

RESUMEN

Covalent DNA-protein cross-links (DPCs) impede replication fork progression and threaten genome integrity. Using Xenopus egg extracts, we previously showed that replication fork collision with DPCs causes their proteolysis, followed by translesion DNA synthesis. We show here that when DPC proteolysis is blocked, the replicative DNA helicase CMG (CDC45, MCM2-7, GINS), which travels on the leading strand template, bypasses an intact leading strand DPC. Single-molecule imaging reveals that GINS does not dissociate from CMG during bypass and that CMG slows dramatically after bypass, likely due to uncoupling from the stalled leading strand. The DNA helicase RTEL1 facilitates bypass, apparently by generating single-stranded DNA beyond the DPC. The absence of RTEL1 impairs DPC proteolysis, suggesting that CMG must bypass the DPC to enable proteolysis. Our results suggest a mechanism that prevents inadvertent CMG destruction by DPC proteases, and they reveal CMG's remarkable capacity to overcome obstacles on its translocation strand.


Asunto(s)
ADN Helicasas/metabolismo , ADN Helicasas/fisiología , Reparación del ADN/fisiología , Animales , Proteínas de Ciclo Celular/metabolismo , ADN/metabolismo , Replicación del ADN , ADN de Cadena Simple , Proteínas de Unión al ADN/fisiología , Femenino , Masculino , Proteolisis , Imagen Individual de Molécula/métodos , Xenopus laevis/metabolismo
2.
Cell ; 176(1-2): 198-212.e15, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30503211

RESUMEN

Understanding transcription factor navigation through the nucleus remains critical for developing targeted therapeutics. The GLI1 transcription factor must maintain maximal Hedgehog pathway output in basal cell carcinomas (BCCs), and we have previously shown that resistant BCCs increase GLI1 deacetylation through atypical protein kinase Cι/λ (aPKC) and HDAC1. Here we identify a lamina-associated polypeptide 2 (LAP2) isoform-dependent nuclear chaperoning system that regulates GLI1 movement between the nuclear lamina and nucleoplasm to achieve maximal activation. LAP2ß forms a two-site interaction with the GLI1 zinc-finger domain and acetylation site, stabilizing an acetylation-dependent reserve on the inner nuclear membrane (INM). By contrast, the nucleoplasmic LAP2α competes with LAP2ß for GLI1 while scaffolding HDAC1 to deacetylate the secondary binding site. aPKC functions to promote GLI1 association with LAP2α, promoting egress off the INM. GLI1 intranuclear trafficking by LAP2 isoforms represents a powerful signal amplifier in BCCs with implications for zinc finger-based signal transduction and therapeutics.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Células 3T3 , Animales , Carcinoma Basocelular/metabolismo , Línea Celular , Cromatina , Proteínas de Unión al ADN/fisiología , Células HEK293 , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiología , Histona Desacetilasa 1/metabolismo , Humanos , Proteínas de la Membrana/fisiología , Ratones , Chaperonas Moleculares/metabolismo , Lámina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteína con Dedos de Zinc GLI1/fisiología , Dedos de Zinc
3.
Cell ; 175(2): 558-570.e11, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30245011

RESUMEN

Given that genomic DNA exerts its function by being transcribed, it is critical for the maintenance of homeostasis that DNA damage, such as double-strand breaks (DSBs), within transcriptionally active regions undergoes accurate repair. However, it remains unclear how this is achieved. Here, we describe a mechanism for transcription-associated homologous recombination repair (TA-HRR) in human cells. The process is initiated by R-loops formed upon DSB induction. We identify Rad52, which is recruited to the DSB site in a DNA-RNA-hybrid-dependent manner, as playing pivotal roles in promoting XPG-mediated R-loop processing and initiating subsequent repair by HRR. Importantly, dysfunction of TA-HRR promotes DSB repair via non-homologous end joining, leading to a striking increase in genomic aberrations. Thus, our data suggest that the presence of R-loops around DSBs within transcriptionally active regions promotes accurate repair of DSBs via processing by Rad52 and XPG to protect genomic information in these critical regions from gene alterations.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Proteínas Nucleares/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Reparación del ADN por Recombinación/fisiología , Factores de Transcripción/metabolismo , Línea Celular , ADN/genética , Roturas del ADN de Doble Cadena , Daño del ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Endonucleasas/fisiología , Recombinación Homóloga , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , ARN/genética , Proteína Recombinante y Reparadora de ADN Rad52/genética , Factores de Transcripción/fisiología
4.
Cell ; 175(2): 442-457.e23, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290143

RESUMEN

Antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) critically contribute to the efficacy of anti-tumor therapeutic antibodies. We report here an unexpected finding that macrophages after ADCP inhibit NK cell-mediated ADCC and T cell-mediated cytotoxicity in breast cancers and lymphomas. Mechanistically, AIM2 is recruited to the phagosomes by FcγR signaling following ADCP and activated by sensing the phagocytosed tumor DNAs through the disrupted phagosomal membrane, which subsequently upregulates PD-L1 and IDO and causes immunosuppression. Combined treatment with anti-HER2 antibody and inhibitors of PD-L1 and IDO enhances anti-tumor immunity and anti-HER2 therapeutic efficacy in mouse models. Furthermore, neoadjuvant trastuzumab therapy significantly upregulates PD-L1 and IDO in the tumor-associated macrophages (TAMs) of HER2+ breast cancer patients, correlating with poor trastuzumab response. Collectively, our findings unveil a deleterious role of ADCP macrophages in cancer immunosuppression and suggest that therapeutic antibody plus immune checkpoint blockade may provide synergistic effects in cancer treatment.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Citofagocitosis/inmunología , Macrófagos/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/fisiología , Antígeno B7-H1/genética , Antígeno B7-H1/fisiología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Citofagocitosis/fisiología , Proteínas de Unión al ADN/fisiología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoterapia , Células Asesinas Naturales/fisiología , Linfoma/inmunología , Macrófagos/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fagocitosis/inmunología , Fagocitosis/fisiología , Fagosomas/fisiología , Receptores de IgG/inmunología
5.
Nat Rev Mol Cell Biol ; 17(7): 399-412, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27075410

RESUMEN

SMC (structural maintenance of chromosomes) complexes - which include condensin, cohesin and the SMC5-SMC6 complex - are major components of chromosomes in all living organisms, from bacteria to humans. These ring-shaped protein machines, which are powered by ATP hydrolysis, topologically encircle DNA. With their ability to hold more than one strand of DNA together, SMC complexes control a plethora of chromosomal activities. Notable among these are chromosome condensation and sister chromatid cohesion. Moreover, SMC complexes have an important role in DNA repair. Recent mechanistic insight into the function and regulation of these universal chromosomal machines enables us to propose molecular models of chromosome structure, dynamics and function, illuminating one of the fundamental entities in biology.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Cromosomas/fisiología , Proteínas de Unión al ADN/fisiología , Complejos Multiproteicos/fisiología , Adenosina Trifosfatasas/ultraestructura , Animales , Ensamble y Desensamble de Cromatina , Cromosomas/ultraestructura , ADN/fisiología , ADN/ultraestructura , Reparación del ADN , Proteínas de Unión al ADN/ultraestructura , Inestabilidad Genómica , Humanos , Complejos Multiproteicos/ultraestructura
6.
Mol Cell ; 77(6): 1222-1236.e13, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32048998

RESUMEN

RNA decay is crucial for mRNA turnover and surveillance and misregulated in many diseases. This complex system is challenging to study, particularly in mammals, where it remains unclear whether decay pathways perform specialized versus redundant roles. Cytoplasmic pathways and links to translation are particularly enigmatic. By directly profiling decay factor targets and normal versus aberrant translation in mouse embryonic stem cells (mESCs), we uncovered extensive decay pathway specialization and crosstalk with translation. XRN1 (5'-3') mediates cytoplasmic bulk mRNA turnover whereas SKIV2L (3'-5') is universally recruited by ribosomes, tackling aberrant translation and sometimes modulating mRNA abundance. Further exploring translation surveillance revealed AVEN and FOCAD as SKIV2L interactors. AVEN prevents ribosome stalls at structured regions, which otherwise require SKIV2L for clearance. This pathway is crucial for histone translation, upstream open reading frame (uORF) regulation, and counteracting ribosome arrest on small ORFs. In summary, we uncovered key targets, components, and functions of mammalian RNA decay pathways and extensive coupling to translation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas de Unión al ADN/fisiología , Exorribonucleasas/fisiología , Células Madre Embrionarias de Ratones/metabolismo , Biosíntesis de Proteínas , ARN Helicasas/fisiología , Estabilidad del ARN , ARN Mensajero/metabolismo , Animales , Sistemas CRISPR-Cas , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología , Sistemas de Lectura Abierta , Proteínas Proto-Oncogénicas/fisiología , ARN Mensajero/química , ARN Mensajero/genética , Ribosomas/genética , Ribosomas/metabolismo
7.
Mol Cell ; 76(5): 724-737.e5, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31629658

RESUMEN

Condensin is a conserved SMC complex that uses its ATPase machinery to structure genomes, but how it does so is largely unknown. We show that condensin's ATPase has a dual role in chromosome condensation. Mutation of one ATPase site impairs condensation, while mutating the second site results in hyperactive condensin that compacts DNA faster than wild-type, both in vivo and in vitro. Whereas one site drives loop formation, the second site is involved in the formation of more stable higher-order Z loop structures. Using hyperactive condensin I, we reveal that condensin II is not intrinsically needed for the shortening of mitotic chromosomes. Condensin II rather is required for a straight chromosomal axis and enables faithful chromosome segregation by counteracting the formation of ultrafine DNA bridges. SMC complexes with distinct roles for each ATPase site likely reflect a universal principle that enables these molecular machines to intricately control chromosome architecture.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Ensamble y Desensamble de Cromatina/fisiología , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , Adenosina Trifosfato/química , Sitios de Unión/genética , Sitios de Unión/fisiología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/metabolismo , Cromosomas/fisiología , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Humanos , Complejos Multiproteicos/fisiología , Unión Proteica/fisiología , Subunidades de Proteína/metabolismo , Cohesinas
8.
Nat Rev Mol Cell Biol ; 15(10): 677-89, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25207437

RESUMEN

Capping protein (CP) binds the fast growing barbed end of the actin filament and regulates actin assembly by blocking the addition and loss of actin subunits. Recent studies provide new insights into how CP and barbed-end capping are regulated. Filament elongation factors, such as formins and ENA/VASP (enabled/vasodilator-stimulated phosphoprotein), indirectly regulate CP by competing with CP for binding to the barbed end, whereas other molecules, including V-1 and phospholipids, directly bind to CP and sterically block its interaction with the filament. In addition, a diverse and unrelated group of proteins interact with CP through a conserved 'capping protein interaction' (CPI) motif. These proteins, including CARMIL (capping protein, ARP2/3 and myosin I linker), CD2AP (CD2-associated protein) and the WASH (WASP and SCAR homologue) complex subunit FAM21, recruit CP to specific subcellular locations and modulate its actin-capping activity via allosteric effects.


Asunto(s)
Proteínas de Capping de la Actina/metabolismo , Citoesqueleto de Actina/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Capping de la Actina/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencia de Aminoácidos , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/fisiología , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/fisiología , Modelos Moleculares , Fosfatos de Fosfatidilinositol/química , Unión Proteica , Conformación Proteica
9.
Annu Rev Cell Dev Biol ; 28: 163-87, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22905954

RESUMEN

Long-range interactions between transcription regulatory elements play an important role in gene activation, epigenetic silencing, and chromatin organization. Transcriptional activation or repression of developmentally regulated genes is often accomplished through tissue-specific chromatin architecture and dynamic localization between active transcription factories and repressive Polycomb bodies. However, the mechanisms underlying the structural organization of chromatin and the coordination of physical interactions are not fully understood. Insulators and Polycomb group proteins form highly conserved multiprotein complexes that mediate functional long-range interactions and have proposed roles in nuclear organization. In this review, we explore recent findings that have broadened our understanding of the function of these proteins and provide an integrative model for the roles of insulators in nuclear organization.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Genoma/fisiología , Animales , Cromatina/metabolismo , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Inestabilidad Genómica , Humanos , Modelos Moleculares , Regiones Promotoras Genéticas , Transcripción Genética
10.
Mol Cell ; 72(3): 583-593.e4, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30293780

RESUMEN

Copy-number changes generate phenotypic variability in health and disease. Whether organisms protect against copy-number changes is largely unknown. Here, we show that Saccharomyces cerevisiae monitors the copy number of its ribosomal DNA (rDNA) and rapidly responds to copy-number loss with the clonal amplification of extrachromosomal rDNA circles (ERCs) from chromosomal repeats. ERC formation is replicative, separable from repeat loss, and reaches a dynamic steady state that responds to the addition of exogenous rDNA copies. ERC levels are also modulated by RNAPI activity and diet, suggesting that rDNA copy number is calibrated against the cellular demand for rRNA. Last, we show that ERCs reinsert into the genome in a dosage-dependent manner, indicating that they provide a reservoir for ultimately increasing rDNA array length. Our results reveal a DNA-based mechanism for rapidly restoring copy number in response to catastrophic gene loss that shares fundamental features with unscheduled copy-number amplifications in cancer cells.


Asunto(s)
Variaciones en el Número de Copia de ADN/fisiología , ADN Circular/fisiología , ADN Ribosómico/fisiología , Variaciones en el Número de Copia de ADN/genética , Replicación del ADN/fisiología , ADN Circular/genética , ADN Circular/metabolismo , ADN Ribosómico/genética , Proteínas de Unión al ADN/fisiología , Genómica , ARN Ribosómico/genética , Recombinación Genética/genética , Ribosomas/fisiología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
11.
Mol Cell ; 72(3): 594-600.e2, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401433

RESUMEN

The +1 nucleosome of yeast genes, within which reside transcription start sites, is characterized by histone acetylation, by the displacement of an H2A-H2B dimer, and by a persistent association with the RSC chromatin-remodeling complex. Here we demonstrate the interrelationship of these characteristics and the conversion of a nucleosome to the +1 state in vitro. Contrary to expectation, acetylation performs an inhibitory role, preventing the removal of a nucleosome by RSC. Inhibition is due to both enhanced RSC-histone interaction and diminished histone-chaperone interaction. Acetylation does not prevent all RSC activity, because stably bound RSC removes an H2A-H2B dimer on a timescale of seconds in an irreversible manner.


Asunto(s)
Ensamble y Desensamble de Cromatina/fisiología , Proteínas de Unión al ADN/fisiología , Histonas/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Factores de Transcripción/fisiología , Acetilcoenzima A/metabolismo , Acetilación , Animales , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Proteína 1 de Ensamblaje de Nucleosomas , Nucleosomas/fisiología , Conformación Proteica , Procesamiento Proteico-Postraduccional , Ratas , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
12.
Genes Dev ; 32(2): 181-193, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29440247

RESUMEN

Epithelial tissues rely on a highly coordinated balance between self-renewal, proliferation, and differentiation, disruption of which may drive carcinogenesis. The epigenetic regulator KMT2D (MLL4) is one of the most frequently mutated genes in all cancers, particularly epithelial cancers, yet its normal function in these tissues is unknown. Here, we identify a novel role for KMT2D in coordinating this fine balance, as depletion of KMT2D from undifferentiated epidermal keratinocytes results in reduced proliferation, premature spurious activation of terminal differentiation genes, and disorganized epidermal stratification. Genome-wide, KMT2D interacts with p63 and is enriched at its target enhancers. Depletion of KMT2D results in a broad loss of enhancer histone modifications H3 Lys 4 (H3K4) monomethylation (H3K4me1) and H3K27 acetylation (H3K27ac) as well as reduced expression of p63 target genes, including key genes involved in epithelial development and adhesion. Together, these results reveal a critical role for KMT2D in the control of epithelial enhancers and p63 target gene expression, including the requirement of KMT2D for the maintenance of epithelial progenitor gene expression and the coordination of proper terminal differentiation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Elementos de Facilitación Genéticos , Queratinocitos/metabolismo , Proteínas de Neoplasias/fisiología , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Código de Histonas , Homeostasis , Humanos , Proteínas de Neoplasias/metabolismo
13.
Genes Dev ; 32(7-8): 524-536, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29636371

RESUMEN

Chromosomal deletion rearrangements mediated by repetitive elements often involve repeats separated by several kilobases and sequences that are divergent. While such rearrangements are likely induced by DNA double-strand breaks (DSBs), it has been unclear how the proximity of DSBs relative to repeat sequences affects the frequency of such events. We generated a reporter assay in mouse cells for a deletion rearrangement involving repeats separated by 0.4 Mb. We induced this repeat-mediated deletion (RMD) rearrangement with two DSBs: the 5' DSB that is just downstream from the first repeat and the 3' DSB that is varying distances upstream of the second repeat. Strikingly, we found that increasing the 3' DSB/repeat distance from 3.3 kb to 28.4 kb causes only a modest decrease in rearrangement frequency. We also found that RMDs are suppressed by KU70 and RAD51 and promoted by RAD52, CtIP, and BRCA1. In addition, we found that 1%-3% sequence divergence substantially suppresses these rearrangements in a manner dependent on the mismatch repair factor MSH2, which is dominant over the suppressive role of KU70. We suggest that a DSB far from a repeat can stimulate repeat-mediated rearrangements, but multiple pathways suppress these events.


Asunto(s)
Rotura Cromosómica , Deleción Cromosómica , Secuencias Repetitivas de Ácidos Nucleicos , Animales , ADN/química , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Autoantígeno Ku/fisiología , Ratones , Proteína 2 Homóloga a MutS/fisiología , Recombinasa Rad51/fisiología , Proteína Recombinante y Reparadora de ADN Rad52/fisiología , Proteína 1 de Unión al Supresor Tumoral P53/fisiología
14.
Genes Dev ; 32(7-8): 537-554, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29654060

RESUMEN

The identification of telomerase-negative HAATI (heterochromatin amplification-mediated and telomerase-independent) cells, in which telomeres are superseded by nontelomeric heterochromatin tracts, challenged the idea that canonical telomeres are essential for chromosome linearity and raised crucial questions as to how such tracts translocate to eroding chromosome ends and confer end protection. Here we show that HAATI arises when telomere loss triggers a newly recognized illegitimate translocation pathway that requires RNAi factors. While RNAi is necessary for the translocation events that mobilize ribosomal DNA (rDNA) tracts to all chromosome ends (forming "HAATIrDNA" chromosomes), it is dispensable for HAATIrDNA maintenance. Surprisingly, Dicer (Dcr1) plays a separate, RNAi-independent role in preventing formation of the rare HAATI subtype in which a different repetitive element (the subtelomeric element) replaces telomeres. Using genetics and fusions between shelterin components and rDNA-binding proteins, we mapped the mechanism by which rDNA loci engage crucial end protection factors-despite the absence of telomere repeats-and secure end protection. Sequence analysis of HAATIrDNA genomes allowed us to propose RNA and DNA polymerase template-switching models for the mechanism of RNAi-triggered rDNA translocations. Collectively, our results reveal unforeseen roles for noncoding RNAs (ncRNAs) in assembling a telomere-free chromosome end protection device.


Asunto(s)
ADN Ribosómico , Heterocromatina , Interferencia de ARN , Translocación Genética , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Recombinasa Rad51/fisiología , Ribonucleasa III/metabolismo , Ribonucleasa III/fisiología , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas de Schizosaccharomyces pombe/fisiología , Complejo Shelterina , Telómero , Proteínas de Unión a Telómeros/metabolismo , Secuencias Repetidas Terminales
15.
Genes Dev ; 32(2): 165-180, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29440260

RESUMEN

Multiple congenital disorders often present complex phenotypes, but how the mutation of individual genetic factors can lead to multiple defects remains poorly understood. In the present study, we used human neuroepithelial (NE) cells and CHARGE patient-derived cells as an in vitro model system to identify the function of chromodomain helicase DNA-binding 7 (CHD7) in NE-neural crest bifurcation, thus revealing an etiological link between the central nervous system (CNS) and craniofacial anomalies observed in CHARGE syndrome. We found that CHD7 is required for epigenetic activation of superenhancers and CNS-specific enhancers, which support the maintenance of the NE and CNS lineage identities. Furthermore, we found that BRN2 and SOX21 are downstream effectors of CHD7, which shapes cellular identities by enhancing a CNS-specific cellular program and indirectly repressing non-CNS-specific cellular programs. Based on our results, CHD7, through its interactions with superenhancer elements, acts as a regulatory hub in the orchestration of the spatiotemporal dynamics of transcription factors to regulate NE and CNS lineage identities.


Asunto(s)
ADN Helicasas/fisiología , Proteínas de Unión al ADN/fisiología , Epigénesis Genética , Células-Madre Neurales/metabolismo , Células Neuroepiteliales/metabolismo , Síndrome CHARGE/genética , Línea Celular , Linaje de la Célula/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Elementos de Facilitación Genéticos , Haploinsuficiencia , Humanos , Cresta Neural/metabolismo , Transcripción Genética
16.
Genes Dev ; 32(1): 70-78, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29386330

RESUMEN

The number of known long noncoding RNA (lncRNA) functions is rapidly growing, but how those functions are encoded in their sequence and structure remains poorly understood. NORAD (noncoding RNA activated by DNA damage) is a recently characterized, abundant, and highly conserved lncRNA that is required for proper mitotic divisions in human cells. NORAD acts in the cytoplasm and antagonizes repressors from the Pumilio family that bind at least 17 sites spread through 12 repetitive units in NORAD sequence. Here we study conserved sequences in NORAD repeats, identify additional interacting partners, and characterize the interaction between NORAD and the RNA-binding protein SAM68 (KHDRBS1), which is required for NORAD function in antagonizing Pumilio. These interactions provide a paradigm for how repeated elements in a lncRNA facilitate function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3' , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Sitios de Unión , Línea Celular Tumoral , Segregación Cromosómica , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Humanos , ARN Largo no Codificante/química , ARN Largo no Codificante/genética , ARN Largo no Codificante/fisiología , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Proteínas Represoras/metabolismo
17.
Nat Immunol ; 14(3): 230-7, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23334790

RESUMEN

Regulatory T cells (T(reg) cells) develop from progenitor thymocytes after the engagement of T cell antigen receptors (TCRs) with high-affinity ligands, but the underlying molecular mechanisms are still unclear. Here we show that the Nr4a nuclear receptors, which are encoded by immediate-early genes upregulated by TCR stimulation in thymocytes, have essential roles in T(reg) cell development. Mice that lacked all Nr4a factors could not produce T(reg) cells and died early owing to systemic autoimmunity. Nr4a receptors directly activated the promoter of the gene encoding the transcription factor Foxp3, and forced activation of Nr4a receptors bypassed low-strength TCR signaling to drive the T(reg) cell developmental program. Our results suggest that Nr4a receptors have key roles in determining CD4(+) T cell fates in the thymus and thus contribute to immune homeostasis.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas del Tejido Nervioso/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Esteroides/fisiología , Receptores de Hormona Tiroidea/fisiología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Autoinmunidad/genética , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Genes Inmediatos-Precoces , Homeostasis , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Esteroides/genética , Receptores de Hormona Tiroidea/genética , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Timocitos/metabolismo
18.
Nat Rev Mol Cell Biol ; 14(4): 197-210, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23847781

RESUMEN

The protein kinase ataxia-telangiectasia mutated (ATM) is best known for its role as an apical activator of the DNA damage response in the face of DNA double-strand breaks (DSBs). Following induction of DSBs, ATM mobilizes one of the most extensive signalling networks that responds to specific stimuli and modifies directly or indirectly a broad range of targets. Although most ATM research has focused on this function, evidence suggests that ATM-mediated phosphorylation has a role in the response to other types of genotoxic stress. Moreover, it has become apparent that ATM is active in other cell signalling pathways involved in maintaining cellular homeostasis.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Daño del ADN , Proteínas de Unión al ADN/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Núcleo Celular/enzimología , Homeostasis , Humanos , Neuronas/enzimología , Oxidación-Reducción , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Vasos Retinianos/enzimología , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
19.
Proc Natl Acad Sci U S A ; 119(37): e2201513119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36067320

RESUMEN

Most animal species display dimorphic sexual behaviors and male-biased aggressiveness. Current models have focused on the male-specific product from the fruitless (fruM) gene, which controls male courtship and male-specific aggression patterns in fruit flies, and describe a male-specific mechanism underlying sexually dimorphic behaviors. Here we show that the doublesex (dsx) gene, which expresses male-specific DsxM and female-specific DsxF transcription factors, functions in the nervous system to control both male and female sexual and aggressive behaviors. We find that Dsx is not only required in central brain neurons for male and female sexual behaviors, but also functions in approximately eight pairs of male-specific neurons to promote male aggressiveness and approximately two pairs of female-specific neurons to inhibit female aggressiveness. DsxF knockdown females fight more frequently, even with males. Our findings reveal crucial roles of dsx, which is broadly conserved from worms to humans, in a small number of neurons in both sexes to establish dimorphic sexual and aggressive behaviors.


Asunto(s)
Agresión , Cortejo , Proteínas de Unión al ADN , Proteínas de Drosophila , Drosophila melanogaster , Conducta Sexual Animal , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Femenino , Masculino
20.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34969846

RESUMEN

The biogenesis of integral ß-barrel outer membrane proteins (OMPs) in gram-negative bacteria requires transport by molecular chaperones across the aqueous periplasmic space. Owing in part to the extensive functional redundancy within the periplasmic chaperone network, specific roles for molecular chaperones in OMP quality control and assembly have remained largely elusive. Here, by deliberately perturbing the OMP assembly process through use of multiple folding-defective substrates, we have identified a role for the periplasmic chaperone Skp in ensuring efficient folding of OMPs by the ß-barrel assembly machine (Bam) complex. We find that ß-barrel substrates that fail to integrate into the membrane in a timely manner are removed from the Bam complex by Skp, thereby allowing for clearance of stalled Bam-OMP complexes. Following the displacement of OMPs from the assembly machinery, Skp subsequently serves as a sacrificial adaptor protein to directly facilitate the degradation of defective OMP substrates by the periplasmic protease DegP. We conclude that Skp acts to ensure efficient ß-barrel folding by directly mediating the displacement and degradation of assembly-compromised OMP substrates from the Bam complex.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas de Escherichia coli/fisiología , Chaperonas Moleculares/fisiología , Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Unión Proteica , Conformación Proteica en Lámina beta , Pliegue de Proteína , Proteolisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA