Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 784
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38862431

RESUMEN

Ribonuclease P (RNase P) was first described in the 1970's as an endoribonuclease acting in the maturation of precursor transfer RNAs (tRNAs). More recent studies, however, have uncovered non-canonical roles for RNase P and its components. Here, we review the recent progress of its involvement in chromatin assembly, DNA damage response, and maintenance of genome stability with implications in tumorigenesis. The possibility of RNase P as a therapeutic target in cancer is also discussed.


Asunto(s)
Neoplasias , Precursores del ARN , ARN de Transferencia , Ribonucleasa P , Ribonucleasa P/metabolismo , Ribonucleasa P/genética , Humanos , ARN de Transferencia/metabolismo , ARN de Transferencia/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/enzimología , Precursores del ARN/metabolismo , Precursores del ARN/genética , Inestabilidad Genómica , Animales , Daño del ADN , Procesamiento Postranscripcional del ARN , Ensamble y Desensamble de Cromatina/genética
2.
Biochem Biophys Res Commun ; 724: 150223, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38852505

RESUMEN

SWI/SNF chromatin remodeling complexes play a key role in gene transcription as epigenetic regulators and are typically considered to act as tumor suppressors in cancers. Compared to other cancer-related components of the SWI/SNF complex, research on SMARCC2, a component of the initial BAF core, has been relatively limited. This study aimed to elucidate the role of SMARCC2 in breast cancer by employing various in vitro and in vivo methods including cell proliferation assays, mammosphere formation, and xenograft models, complemented by RNA-seq, ATAC-seq, and ChIP analyses. The results showed that SMARCC2 silencing surprisingly led to the suppression of breast tumorigenesis, indicating a pro-tumorigenic function for SMARCC2 in breast cancer, which contrasts with the roles of other SWI/SNF subunits. In addition, SMARCC2 depletion reduces cancer stem cell features of breast cancer cells. Mechanistic study showed that SMARCC2 silencing downregulated the oncogenic Ras-PI3K signaling pathway, likely by directly regulating the chromatin accessibility of the enhancers of the key genes such as PIK3CB. Together, these results expand our understanding of the SWI/SNF complex's role in cancer development and identify SMARCC2 as a promising new target for breast cancer therapies.


Asunto(s)
Neoplasias de la Mama , Cromatina , Silenciador del Gen , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Cromatina/metabolismo , Cromatina/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proliferación Celular/genética , Carcinogénesis/genética , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Transducción de Señal , Ratones Desnudos , Ensamble y Desensamble de Cromatina/genética
3.
Nat Commun ; 15(1): 4770, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839769

RESUMEN

SMARCB1, a subunit of the SWI/SNF chromatin remodeling complex, is the causative gene of rhabdoid tumors and epithelioid sarcomas. Here, we identify a paralog pair of CBP and p300 as a synthetic lethal target in SMARCB1-deficient cancers by using a dual siRNA screening method based on the "simultaneous inhibition of a paralog pair" concept. Treatment with CBP/p300 dual inhibitors suppresses growth of cell lines and tumor xenografts derived from SMARCB1-deficient cells but not from SMARCB1-proficient cells. SMARCB1-containing SWI/SNF complexes localize with H3K27me3 and its methyltransferase EZH2 at the promotor region of the KREMEN2 locus, resulting in transcriptional downregulation of KREMEN2. By contrast, SMARCB1 deficiency leads to localization of H3K27ac, and recruitment of its acetyltransferases CBP and p300, at the KREMEN2 locus, resulting in transcriptional upregulation of KREMEN2, which cooperates with the SMARCA1 chromatin remodeling complex. Simultaneous inhibition of CBP/p300 leads to transcriptional downregulation of KREMEN2, followed by apoptosis induction via monomerization of KREMEN1 due to a failure to interact with KREMEN2, which suppresses anti-apoptotic signaling pathways. Taken together, our findings indicate that simultaneous inhibitors of CBP/p300 could be promising therapeutic agents for SMARCB1-deficient cancers.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteína SMARCB1 , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo , Humanos , Animales , Línea Celular Tumoral , Ratones , Factores de Transcripción p300-CBP/metabolismo , Factores de Transcripción p300-CBP/genética , Proteína p300 Asociada a E1A/metabolismo , Proteína p300 Asociada a E1A/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Ensamble y Desensamble de Cromatina/genética , Ratones Desnudos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Regiones Promotoras Genéticas/genética , Proliferación Celular/genética , Proliferación Celular/efectos de los fármacos , Tumor Rabdoide/genética , Tumor Rabdoide/metabolismo , Tumor Rabdoide/patología
4.
Genes (Basel) ; 15(5)2024 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-38790189

RESUMEN

BACKGROUND: Cervical cancer is among the highest-ranking types of cancer worldwide, with human papillomavirus (HPV) as the agent driving the malignant process. One aspect of the infection's evolution is given by epigenetic modifications, mainly DNA methylation and chromatin alteration. These processes are guided by several chromatin remodeling complexes, including NuRD. The purpose of this study was to evaluate the genome-wide binding patterns of the NuRD complex components (MBD2 and MBD3) in the presence of active HPV16 E6 and E7 oncogenes and to determine the potential of identified genes through an experimental model to differentiate between cervical precursor lesions, with the aim of establishing their utility as biomarkers. METHODS: The experimental model was built using the CaSki cell line and shRNA for E6 and E7 HPV16 silencing, ChIP-seq, qRT-PCR, and Western blot analyses. Selected genes' expression was also assessed in patients. RESULTS: Several genes have been identified to exhibit altered transcriptional activity due to the influence of HPV16 E6/E7 viral oncogenes acting through the MBD2/MBD3 NuRD complex, linking them to viral infection and cervical oncogenesis. CONCLUSIONS: The impacted genes primarily play roles in governing gene transcription, mRNA processing, and regulation of translation. Understanding these mechanisms offers valuable insights into the process of HPV-induced oncogenesis.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN , Papillomavirus Humano 16 , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2 , Proteínas Oncogénicas Virales , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Femenino , Humanos , Carcinogénesis/genética , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 16/patogenicidad , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Neoplasias del Cuello Uterino/virología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
5.
Artículo en Inglés | MEDLINE | ID: mdl-38761362

RESUMEN

Epigenetic changes have been established to be a hallmark of aging, which implies that aging science requires collaborating with the field of chromatin biology. DNA methylation patterns, changes in relative abundance of histone post-translational modifications, and chromatin remodeling are the central players in modifying chromatin structure. Aging is commonly associated with an overall increase in chromatin instability, loss of homeostasis, and decondensation. However, numerous publications have highlighted that the link between aging and chromatin changes is not nearly as linear as previously expected. This complex interplay of these epigenetic elements during the lifetime of an organism likely contributes to cellular senescence, genomic instability, and disease susceptibility. Yet, the causal links between these phenomena still need to be fully unraveled. In this perspective article, we discuss potential future directions of aging chromatin biology.


Asunto(s)
Envejecimiento , Cromatina , Epigénesis Genética , Neoplasias , Humanos , Envejecimiento/genética , Envejecimiento/fisiología , Cromatina/genética , Cromatina/metabolismo , Neoplasias/genética , Senescencia Celular/genética , Senescencia Celular/fisiología , Inestabilidad Genómica/genética , Ensamble y Desensamble de Cromatina/genética , Metilación de ADN , Histonas/metabolismo , Animales , Procesamiento Proteico-Postraduccional
6.
Nucleic Acids Res ; 52(10): 5698-5719, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38587186

RESUMEN

AT-rich interaction domain protein 1A (ARID1A), a SWI/SNF chromatin remodeling complex subunit, is frequently mutated across various cancer entities. Loss of ARID1A leads to DNA repair defects. Here, we show that ARID1A plays epigenetic roles to promote both DNA double-strand breaks (DSBs) repair pathways, non-homologous end-joining (NHEJ) and homologous recombination (HR). ARID1A is accumulated at DSBs after DNA damage and regulates chromatin loops formation by recruiting RAD21 and CTCF to DSBs. Simultaneously, ARID1A facilitates transcription silencing at DSBs in transcriptionally active chromatin by recruiting HDAC1 and RSF1 to control the distribution of activating histone marks, chromatin accessibility, and eviction of RNAPII. ARID1A depletion resulted in enhanced accumulation of micronuclei, activation of cGAS-STING pathway, and an increased expression of immunomodulatory cytokines upon ionizing radiation. Furthermore, low ARID1A expression in cancer patients receiving radiotherapy was associated with higher infiltration of several immune cells. The high mutation rate of ARID1A in various cancer types highlights its clinical relevance as a promising biomarker that correlates with the level of immune regulatory cytokines and estimates the levels of tumor-infiltrating immune cells, which can predict the response to the combination of radio- and immunotherapy.


Asunto(s)
Cromatina , Reparación del ADN , Proteínas de Unión al ADN , Inmunidad , Factores de Transcripción , Humanos , Línea Celular Tumoral , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Recombinación Homóloga/genética , Inmunidad/genética , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/inmunología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transactivadores , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Cell Rep Med ; 5(5): 101510, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38614093

RESUMEN

Key gene mutations are essential for colorectal cancer (CRC) development; however, how the mutated tumor cells impact the surrounding normal cells to promote tumor progression has not been well defined. Here, we report that PIK3CA mutant tumor cells transmit oncogenic signals and result in malignant transformation of intestinal epithelial cells (IECs) via paracrine exosomal arachidonic acid (AA)-induced H3K4 trimethylation. Mechanistically, PIK3CA mutations sustain SGK3-FBW7-mediated stability of the cPLA2 protein, leading to the synthetic increase in AA, which is transported through exosome and accumulated in IECs. Transferred AA directly binds Menin and strengthens the interactions of Menin and MLL1/2 methyltransferase. Finally, the combination of VTP50469, an inhibitor of the Menin-MLL interaction, and alpelisib synergistically represses PDX tumors harboring PIK3CA mutations. Together, these findings unveil the metabolic link between PIK3CA mutant tumor cells and the IECs, highlighting AA as the potential target for the treatment of patients with CRC harboring PIK3CA mutations.


Asunto(s)
Ácido Araquidónico , Transformación Celular Neoplásica , Ensamble y Desensamble de Cromatina , Fosfatidilinositol 3-Quinasa Clase I , Mutación , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Ácido Araquidónico/metabolismo , Animales , Mutación/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ensamble y Desensamble de Cromatina/genética , Ratones , Línea Celular Tumoral , Colon/patología , Colon/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Exosomas/metabolismo , Exosomas/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Histonas/metabolismo , Histonas/genética
8.
Adv Sci (Weinh) ; 11(16): e2303379, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38380561

RESUMEN

Patient-Derived Organoids (PDO) and Xenografts (PDX) are the current gold standards for patient-derived models of cancer (PDMC). Nevertheless, how patient tumor cells evolve in these models and the impact on drug response remains unclear. Herein, the transcriptomic and chromatin accessibility landscapes of matched colorectal cancer (CRC) PDO, PDX, PDO-derived PDX (PDOX), and original patient tumors (PT) are compared. Two major remodeling axes are discovered. The first axis delineates PDMC from PT, and the second axis distinguishes PDX and PDO. PDOX are more similar to PDX than PDO, indicating the growth environment is a driving force for chromatin adaptation. Transcription factors (TF) that differentially bind to open chromatins between matched PDO and PDOX are identified. Among them, KLF14 and EGR2 footprints are enriched in PDOX relative to matched PDO, and silencing of KLF14 or EGR2 promoted tumor growth. Furthermore, EPHA4, a shared downstream target gene of KLF14 and EGR2, altered tumor sensitivity to MEK inhibitor treatment. Altogether, patient-derived CRC cells undergo both common and distinct chromatin remodeling in PDO and PDX/PDOX, driven largely by their respective microenvironments, which results in differences in growth and drug sensitivity and needs to be taken into consideration when interpreting their ability to predict clinical outcome.


Asunto(s)
Ensamble y Desensamble de Cromatina , Neoplasias Colorrectales , Organoides , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Humanos , Ensamble y Desensamble de Cromatina/genética , Ratones , Animales , Organoides/metabolismo , Modelos Animales de Enfermedad
9.
Nucleic Acids Res ; 52(4): 1527-1543, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38272542

RESUMEN

The NF-κB protein p65/RelA plays a pivotal role in coordinating gene expression in response to diverse stimuli, including viral infections. At the chromatin level, p65/RelA regulates gene transcription and alternative splicing through promoter enrichment and genomic exon occupancy, respectively. The intricate ways in which p65/RelA simultaneously governs these functions across various genes remain to be fully elucidated. In this study, we employed the HTLV-1 Tax oncoprotein, a potent activator of NF-κB, to investigate its influence on the three-dimensional organization of the genome, a key factor in gene regulation. We discovered that Tax restructures the 3D genomic landscape, bringing together genes based on their regulation and splicing patterns. Notably, we found that the Tax-induced gene-gene contact between the two master genes NFKBIA and RELA is associated with their respective changes in gene expression and alternative splicing. Through dCas9-mediated approaches, we demonstrated that NFKBIA-RELA interaction is required for alternative splicing regulation and is caused by an intragenic enrichment of p65/RelA on RELA. Our findings shed light on new regulatory mechanisms upon HTLV-1 Tax and underscore the integral role of p65/RelA in coordinated regulation of NF-κB-responsive genes at both transcriptional and splicing levels in the context of the 3D genome.


The NF-κB pathway is essential for coordinating gene expression in response to various stimuli, including viral infections. Most studies have focused on the role of NF-κB in transcriptional regulation. In the present study, the impact of the potent NF-κB activator HTLV-1 Tax oncoprotein on the three-dimensional organization of the genome was investigated. Tax-mediated NF-κB activation was found to restructure the 3D genomic landscape in cells and to bring genes together in multigene complexes that are coordinately regulated either transcriptionally or through alternative splicing by NF-κB. Induced coordinate changes in transcription and alternative splicing included the two master genes of NF-κB pathway NFKBIA and RELA. The findings have significant implications for understanding cell fate determination and disease development associated with HTLV-1 infection, as well as chronic NF-κB activation in various human inflammatory diseases and cancer.


Asunto(s)
Ensamble y Desensamble de Cromatina , Regulación de la Expresión Génica , Subunidad p50 de NF-kappa B , Empalme Alternativo/genética , Ensamble y Desensamble de Cromatina/genética , Productos del Gen tax/genética , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Transducción de Señal , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Activación Transcripcional , Humanos , Subunidad p50 de NF-kappa B/metabolismo
10.
Nat Genet ; 56(1): 100-111, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38049663

RESUMEN

Chromatin accessibility is a hallmark of active transcription and entails ATP-dependent nucleosome remodeling, which is carried out by complexes such as Brahma-associated factor (BAF). However, the mechanistic links between transcription, nucleosome remodeling and chromatin accessibility are unclear. Here, we used a chemical-genetic approach coupled with time-resolved chromatin profiling to dissect the interplay between RNA Polymerase II (RNAPII), BAF and DNA-sequence-specific transcription factors in mouse embryonic stem cells. We show that BAF dynamically unwraps and evicts nucleosomes at accessible chromatin regions, while RNAPII promoter-proximal pausing stabilizes BAF chromatin occupancy and enhances ATP-dependent nucleosome eviction by BAF. We find that although RNAPII and BAF dynamically probe both transcriptionally active and Polycomb-repressed genomic regions, pluripotency transcription factor chromatin binding confers locus specificity for productive chromatin remodeling and nucleosome eviction by BAF. Our study suggests a paradigm for how functional synergy between dynamically acting chromatin factors regulates locus-specific nucleosome organization and chromatin accessibility.


Asunto(s)
Nucleosomas , Factores de Transcripción , Animales , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Nucleosomas/genética , Cromatina/genética , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , Ensamble y Desensamble de Cromatina/genética , Adenosina Trifosfato
11.
Nucleic Acids Res ; 52(1): 337-354, 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38000389

RESUMEN

Baz2B is a regulatory subunit of the ATP-dependent chromatin remodeling complexes BRF1 and BRF5, which control access to DNA during DNA-templated processes. Baz2B has been implicated in several diseases and also in unhealthy ageing, however limited information is available on the domains and cellular roles of Baz2B. To gain more insight into the Baz2B function, we biochemically characterized the TAM (Tip5/ARBP/MBD) domain with the auxiliary AT-hook motifs and the bromodomain (BRD). We observed alterations in histone code recognition in bromodomains carrying cancer-associated point mutations, suggesting their potential involvement in disease. Furthermore, the depletion of Baz2B in the Hap1 cell line resulted in altered cell morphology, reduced colony formation and perturbed transcriptional profiles. Despite that, super-resolution microscopy images revealed no changes in the overall chromatin structure in the absence of Baz2B. These findings provide insights into the biological function of Baz2B.


Asunto(s)
Ensamble y Desensamble de Cromatina , Factores de Transcripción , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , ADN , Dominios Proteicos , Factores de Transcripción/genética , Humanos
12.
Braz J Med Biol Res ; 56: e12854, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37970920

RESUMEN

During the tumorigenic process, cancer cells may become overly dependent on the activity of backup cellular pathways for their survival, representing vulnerabilities that could be exploited as therapeutic targets. Certain molecular vulnerabilities manifest as a synthetic lethality relationship, and the identification and characterization of new synthetic lethal interactions may pave the way for the development of new therapeutic approaches for human cancer. Our goal was to investigate a possible synthetic lethal interaction between a member of the Chromodomain Helicase DNA binding proteins family (CHD4) and a member of the histone methyltransferases family (SETDB1) in the molecular context of a cell line (Hs578T) representing the triple negative breast cancer (TNBC), a subtype of breast cancer lacking validated molecular targets for treatment. Therefore, we employed the CRISPR-Cas9 gene editing tool to individually or simultaneously introduce indels in the genomic loci corresponding to the catalytic domains of SETDB1 and CHD4 in the Hs578T cell line. Our main findings included: a) introduction of indels in exon 22 of SETDB1 sensitized Hs578T to the action of the genotoxic chemotherapy doxorubicin; b) by sequentially introducing indels in exon 22 of SETDB1 and exon 23 of CHD4 and tracking the percentage of the remaining wild-type sequences in the mixed cell populations generated, we obtained evidence of the existence of a synthetic lethality interaction between these genes. Considering the lack of molecular targets in TNBC, our findings provided valuable insights for development of new therapeutic approaches not only for TNBC but also for other cancer types.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Histona Metiltransferasas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Ensamble y Desensamble de Cromatina/genética , Mutaciones Letales Sintéticas/genética , Línea Celular , Factores de Transcripción/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo
13.
Psychiatr Genet ; 33(6): 213-232, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37851134

RESUMEN

Chromatin, a protein-DNA complex, is a dynamic structure that stores genetic information within the nucleus and responds to molecular/cellular changes in its structure, providing conditional access to the genetic machinery. ATP-dependent chromatin modifiers regulate access of transcription factors and RNA polymerases to DNA by either "opening" or "closing" the structure of chromatin, and its aberrant regulation leads to a variety of neurodevelopmental disorders. The chromodomain helicase DNA-binding (CHD) proteins are ATP-dependent chromatin modifiers involved in the organization of chromatin structure, act as gatekeepers of genomic access, and deposit histone variants required for gene regulation. In this review, we first discuss the structural and functional domains of the CHD proteins, and their binding sites, and phosphorylation, acetylation, and methylation sites. The conservation of important amino acids in SWItch/sucrose non-fermenting (SWI/SNF) domains, and their protein and mRNA tissue expression profiles are discussed. Next, we convey the important binding partners of CHD proteins, their protein complexes and activities, and their involvements in epigenetic regulation. We also show the ChIP-seq binding dynamics for CHD1, CHD2, CHD4, and CHD7 proteins at promoter regions of histone genes, as well as several genes that are critical for neurodevelopment. The role of CHD proteins in development is also discussed. Finally, this review provides information about CHD protein mutations reported in autism and neurodevelopmental disorders, and their pathogenicity. Overall, this review provides information on the progress of research into CHD proteins, their structural and functional domains, epigenetics, and their role in stem cell, development, and neurological disorders.


Asunto(s)
Trastorno Autístico , Enfermedades del Sistema Nervioso , Humanos , Cromatina/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histonas/genética , Histonas/metabolismo , Epigénesis Genética , Trastorno Autístico/genética , Ensamble y Desensamble de Cromatina/genética , ADN , ADN Helicasas/genética , ADN Helicasas/química , ADN Helicasas/metabolismo , Adenosina Trifosfato/metabolismo , Enfermedades del Sistema Nervioso/genética
14.
BMC Cancer ; 23(1): 811, 2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37648998

RESUMEN

Cancer is one of the main causes of human death. Here, we focus on the B-cell lymphoma 7 protein family member B (BCL7B) gene, an accessory subunit of the SWI/SNF chromatin-remodelling complex. To characterize the function of BCL7B, heterozygous BCL7B-deficient stomach cancer cell lines were generated with the CRISPR/Cas9 genome editing system. The comprehensive gene expression patterns were compared between parental cells and each ΔBCL7B cell line by RNA-seq. The results showed marked downregulation of immune-related genes and upregulation of stemness-related genes in the ΔBCL7B cell lines. Moreover, by ChIP-seq analysis with H3K27me3 antibody, the changes of epigenetic modification sequences were compared between parental cells and each ΔBCL7B cell line. After machine learning, we detected the centroid sequence changes, which exerted an impact on antigen presentation. The regulation of BCL7B expression in cancer cells gives rise to cancer stem cell-like characteristics and the acquisition of an immune evasion phenotype.


Asunto(s)
Neoplasias Gástricas , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Anticuerpos , Línea Celular , Ensamble y Desensamble de Cromatina/genética , Proteínas
15.
Nucleic Acids Res ; 51(19): 10278-10291, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37650639

RESUMEN

In eukaryotes, ATP-dependent chromatin remodelers regulate gene expression in response to nutritional and metabolic stimuli. However, altered transcription of metabolic genes may have significant indirect consequences which are currently poorly understood. In this study, we use genetic and molecular approaches to uncover a role for the remodeler Swi-Snf as a critical regulator of metabolism. We find that snfΔ mutants display a cysteine-deficient phenotype, despite growth in nutrient-rich media. This correlates with widespread perturbations in sulfur metabolic gene transcription, including global redistribution of the sulfur-sensing transcription factor Met4. Our findings show how a chromatin remodeler can have a significant impact on a whole metabolic pathway by directly regulating an important gene subset and demonstrate an emerging role for chromatin remodeling complexes as decisive factors in metabolic control.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona , Ensamble y Desensamble de Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromatina/genética , Expresión Génica , Azufre/metabolismo
16.
Nucleic Acids Res ; 51(17): 9055-9074, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37470997

RESUMEN

The SWI/SNF family of ATP-dependent chromatin remodeling complexes is implicated in multiple DNA damage response mechanisms and frequently mutated in cancer. The BAF, PBAF and ncBAF complexes are three major types of SWI/SNF complexes that are functionally distinguished by their exclusive subunits. Accumulating evidence suggests that double-strand breaks (DSBs) in transcriptionally active DNA are preferentially repaired by a dedicated homologous recombination pathway. We show that different BAF, PBAF and ncBAF subunits promote homologous recombination and are rapidly recruited to DSBs in a transcription-dependent manner. The PBAF and ncBAF complexes promote RNA polymerase II eviction near DNA damage to rapidly initiate transcriptional silencing, while the BAF complex helps to maintain this transcriptional silencing. Furthermore, ARID1A-containing BAF complexes promote RNaseH1 and RAD52 recruitment to facilitate R-loop resolution and DNA repair. Our results highlight how multiple SWI/SNF complexes perform different functions to enable DNA repair in the context of actively transcribed genes.


Asunto(s)
Proteínas Cromosómicas no Histona , Estructuras R-Loop , Ensamble y Desensamble de Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN , Reparación del ADN/genética , Recombinación Homóloga/genética , Humanos
17.
Nat Genet ; 55(8): 1400-1412, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37500730

RESUMEN

DNA sequencing-based studies of neurodevelopmental disorders (NDDs) have identified a wide range of genetic determinants. However, a comprehensive analysis of these data, in aggregate, has not to date been performed. Here, we find that genes encoding the mammalian SWI/SNF (mSWI/SNF or BAF) family of ATP-dependent chromatin remodeling protein complexes harbor the greatest number of de novo missense and protein-truncating variants among nuclear protein complexes. Non-truncating NDD-associated protein variants predominantly disrupt the cBAF subcomplex and cluster in four key structural regions associated with high disease severity, including mSWI/SNF-nucleosome interfaces, the ATPase-core ARID-armadillo repeat (ARM) module insertion site, the Arp module and DNA-binding domains. Although over 70% of the residues perturbed in NDDs overlap with those mutated in cancer, ~60% of amino acid changes are NDD-specific. These findings provide a foundation to functionally group variants and link complex aberrancies to phenotypic severity, serving as a resource for the chromatin, clinical genetics and neurodevelopment communities.


Asunto(s)
Ensamble y Desensamble de Cromatina , Trastornos del Neurodesarrollo , Animales , Humanos , Ensamble y Desensamble de Cromatina/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Cromatina/genética , Nucleosomas , Trastornos del Neurodesarrollo/genética , Mamíferos/genética
18.
Development ; 150(13)2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-37294080

RESUMEN

Coordinated spatio-temporal regulation of the determination and differentiation of neural stem cells is essential for brain development. Failure to integrate multiple factors leads to defective brain structures or tumour formation. Previous studies suggest changes of chromatin state are needed to direct neural stem cell differentiation, but the mechanisms are unclear. Analysis of Snr1, the Drosophila orthologue of SMARCB1, an ATP-dependent chromatin remodelling protein, identified a key role in regulating the transition of neuroepithelial cells into neural stem cells and subsequent differentiation of neural stem cells into the cells needed to build the brain. Loss of Snr1 in neuroepithelial cells leads to premature neural stem cell formation. Additionally, loss of Snr1 in neural stem cells results in inappropriate perdurance of neural stem cells into adulthood. Snr1 reduction in neuroepithelial or neural stem cells leads to the differential expression of target genes. We find that Snr1 is associated with the actively transcribed chromatin region of these target genes. Thus, Snr1 likely regulates the chromatin state in neuroepithelial cells and maintains chromatin state in neural stem cells for proper brain development.


Asunto(s)
Proteínas de Drosophila , Factores de Transcripción , Animales , Factores de Transcripción/metabolismo , Transactivadores/genética , Ensamble y Desensamble de Cromatina/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/metabolismo , Diferenciación Celular/genética , Cromatina
19.
Sci Adv ; 9(17): eade8184, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37115930

RESUMEN

Rhabdomyosarcoma (RMS) is a common soft tissue sarcoma in children that resembles developing skeletal muscle. Unlike normal muscle cells, RMS cells fail to differentiate despite expression of the myogenic determination protein MYOD. The TWIST2 transcription factor is frequently overexpressed in fusion-negative RMS (FN-RMS). TWIST2 blocks differentiation by inhibiting MYOD activity in myoblasts, but its role in FN-RMS pathogenesis is incompletely understood. Here, we show that knockdown of TWIST2 enables FN-RMS cells to exit the cell cycle and undergo terminal myogenesis. TWIST2 knockdown also substantially reduces tumor growth in a mouse xenograft model of FN-RMS. Mechanistically, TWIST2 controls H3K27 acetylation at distal enhancers by interacting with the chromatin remodelers SMARCA4 and CHD3 to activate growth-related target genes and repress myogenesis-related target genes. These findings provide insights into the role of TWIST2 in maintaining an undifferentiated and tumorigenic state of FN-RMS and highlight the potential of suppressing TWIST2-regulated pathways to treat FN-RMS.


Asunto(s)
Rabdomiosarcoma , Sarcoma , Humanos , Animales , Ratones , Ensamble y Desensamble de Cromatina/genética , Regulación Neoplásica de la Expresión Génica , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Sarcoma/genética , Diferenciación Celular/genética , Línea Celular Tumoral , ADN Helicasas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Represoras/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
20.
Nucleic Acids Res ; 51(12): 5981-5996, 2023 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-37099375

RESUMEN

Progesterone receptor (PGR) plays diverse roles in reproductive tissues and thus coordinates mammalian fertility. In the ovary, rapid acute induction of PGR is the key determinant of ovulation through transcriptional control of a unique set of genes that culminates in follicle rupture. However, the molecular mechanisms for this specialized PGR function in ovulation is poorly understood. We have assembled a detailed genomic profile of PGR action through combined ATAC-seq, RNA-seq and ChIP-seq analysis in wildtype and isoform-specific PGR null mice. We demonstrate that stimulating ovulation rapidly reprograms chromatin accessibility in two-thirds of sites, correlating with altered gene expression. An ovary-specific PGR action involving interaction with RUNX transcription factors was observed with 70% of PGR-bound regions also bound by RUNX1. These transcriptional complexes direct PGR binding to proximal promoter regions. Additionally, direct PGR binding to the canonical NR3C motif enable chromatin accessibility. Together these PGR actions mediate induction of essential ovulatory genes. Our findings highlight a novel PGR transcriptional mechanism specific to ovulation, providing new targets for infertility treatments or new contraceptives that block ovulation.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Regulación de la Expresión Génica , Receptores de Progesterona , Transcripción Genética , Animales , Femenino , Ratones , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Mamíferos/genética , Ratones Noqueados , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA