Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 177(3): 608-621.e12, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30955891

RESUMEN

Normal tissues accumulate genetic changes with age, but it is unknown if somatic mutations promote clonal expansion of non-malignant cells in the setting of chronic degenerative diseases. Exome sequencing of diseased liver samples from 82 patients revealed a complex mutational landscape in cirrhosis. Additional ultra-deep sequencing identified recurrent mutations in PKD1, PPARGC1B, KMT2D, and ARID1A. The number and size of mutant clones increased as a function of fibrosis stage and tissue damage. To interrogate the functional impact of mutated genes, a pooled in vivo CRISPR screening approach was established. In agreement with sequencing results, examination of 147 genes again revealed that loss of Pkd1, Kmt2d, and Arid1a promoted clonal expansion. Conditional heterozygous deletion of these genes in mice was also hepatoprotective in injury assays. Pre-malignant somatic alterations are often viewed through the lens of cancer, but we show that mutations can promote regeneration, likely independent of carcinogenesis.


Asunto(s)
Hepatopatías/patología , Hígado/metabolismo , Regeneración , Animales , Enfermedad Crónica , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Hidrolasas/deficiencia , Hidrolasas/genética , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Hepatopatías/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regeneración/fisiología , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuenciación del Exoma
2.
Nat Immunol ; 18(9): 1035-1045, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28759003

RESUMEN

MLL4 is an essential subunit of the histone H3 Lys4 (H3K4)-methylation complexes. We found that MLL4 deficiency compromised the development of regulatory T cells (Treg cells) and resulted in a substantial decrease in monomethylated H3K4 (H3K4me1) and chromatin interaction at putative gene enhancers, a considerable portion of which were not direct targets of MLL4 but were enhancers that interacted with MLL4-bound sites. The decrease in H3K4me1 and chromatin interaction at the enhancers not bound by MLL4 correlated with MLL4 binding at distant interacting regions. Deletion of an upstream MLL4-binding site diminished the abundance of H3K4me1 at the regulatory elements of the gene encoding the transcription factor Foxp3 that were looped to the MLL4-binding site and compromised both the thymic differentiation and the inducible differentiation of Treg cells. We found that MLL4 catalyzed methylation of H3K4 at distant unbound enhancers via chromatin looping, which identifies a previously unknown mechanism for regulating the T cell enhancer landscape and affecting Treg cell differentiation.


Asunto(s)
Diferenciación Celular/genética , Cromatina/metabolismo , Factores de Transcripción Forkhead/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Linfocitos T Reguladores , Animales , Sistemas CRISPR-Cas , Citocinas/inmunología , Citometría de Flujo , Regulación de la Expresión Génica , Immunoblotting , Técnicas In Vitro , Metilación , Ratones
3.
Genes Dev ; 35(9-10): 713-728, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33888555

RESUMEN

MED1 often serves as a surrogate of the general transcription coactivator complex Mediator for identifying active enhancers. MED1 is required for phenotypic conversion of fibroblasts to adipocytes in vitro, but its role in adipose development and expansion in vivo has not been reported. Here, we show that MED1 is not generally required for transcription during adipogenesis in culture and that MED1 is dispensable for adipose development in mice. Instead, MED1 is required for postnatal adipose expansion and the induction of fatty acid and triglyceride synthesis genes after pups switch diet from high-fat maternal milk to carbohydrate-based chow. During adipogenesis, MED1 is dispensable for induction of lineage-determining transcription factors (TFs) PPARγ and C/EBPα but is required for lipid accumulation in the late phase of differentiation. Mechanistically, MED1 controls the induction of lipogenesis genes by facilitating lipogenic TF ChREBP- and SREBP1a-dependent recruitment of Mediator to active enhancers. Together, our findings identify a cell- and gene-specific regulatory role of MED1 as a lipogenesis coactivator required for postnatal adipose expansion.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/genética , Lipogénesis/genética , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/embriología , Animales , Células Cultivadas , Dieta , Ratones , Unión Proteica/genética
4.
Cell ; 153(6): 1266-80, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23727112

RESUMEN

The DNA damage response (DDR) protein 53BP1 protects DNA ends from excessive resection in G1, and thereby favors repair by nonhomologous end-joining (NHEJ) as opposed to homologous recombination (HR). During S phase, BRCA1 antagonizes 53BP1 to promote HR. The pro-NHEJ and antirecombinase functions of 53BP1 are mediated in part by RIF1, the only known factor that requires 53BP1 phosphorylation for its recruitment to double-strand breaks (DSBs). Here, we show that a 53BP1 phosphomutant, 53BP18A, comprising alanine substitutions of the eight most N-terminal S/TQ phosphorylation sites, mimics 53BP1 deficiency by restoring genome stability in BRCA1-deficient cells yet behaves like wild-type 53BP1 with respect to immunoglobulin class switch recombination (CSR). 53BP18A recruits RIF1 but fails to recruit the DDR protein PTIP to DSBs, and disruption of PTIP phenocopies 53BP18A. We conclude that 53BP1 promotes productive CSR and suppresses mutagenic DNA repair through distinct phosphodependent interactions with RIF1 and PTIP.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Cambio de Clase de Inmunoglobulina , Proteínas Nucleares/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Animales , Linfocitos B/metabolismo , Proteína BRCA1/metabolismo , Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Inestabilidad Genómica , Ratones , Mutación , Proteína 1 de Unión al Supresor Tumoral P53
5.
Mol Cell ; 78(6): 1114-1132.e10, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32446320

RESUMEN

Bromodomain-containing protein 4 (BRD4) is a cancer therapeutic target in ongoing clinical trials disrupting primarily BRD4-regulated transcription programs. The role of BRD4 in cancer has been attributed mainly to the abundant long isoform (BRD4-L). Here we show, by isoform-specific knockdown and endogenous protein detection, along with transgene expression, the less abundant BRD4 short isoform (BRD4-S) is oncogenic while BRD4-L is tumor-suppressive in breast cancer cell proliferation and migration, as well as mammary tumor formation and metastasis. Through integrated RNA-seq, genome-wide ChIP-seq, and CUT&RUN association profiling, we identify the Engrailed-1 (EN1) homeobox transcription factor as a key BRD4-S coregulator, particularly in triple-negative breast cancer. BRD4-S and EN1 comodulate the extracellular matrix (ECM)-associated matrisome network, including type II cystatin gene cluster, mucin 5, and cathepsin loci, via enhancer regulation of cancer-associated genes and pathways. Our work highlights the importance of targeted therapies for the oncogenic, but not tumor-suppressive, activity of BRD4.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Genes Homeobox , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Transcripción Genética/genética , Neoplasias de la Mama Triple Negativas/genética
6.
Development ; 151(2)2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38063851

RESUMEN

Cornelia de Lange syndrome (CdLS) is a congenital disorder featuring facial dysmorphism, postnatal growth deficits, cognitive disability and upper limb abnormalities. CdLS is genetically heterogeneous, with cases arising from mutation of BRD4, a bromodomain protein that binds and reads acetylated histones. In this study, we have modeled CdLS facial pathology through mouse neural crest cell (NCC)-specific mutation of BRD4 to characterize cellular and molecular function in craniofacial development. Mice with BRD4 NCC loss of function died at birth with severe facial hypoplasia, cleft palate, mid-facial clefting and exencephaly. Following migration, BRD4 mutant NCCs initiated RUNX2 expression for differentiation to osteoblast lineages but failed to induce downstream RUNX2 targets required for lineage commitment. BRD4 bound to active enhancers to regulate expression of osteogenic transcription factors and extracellular matrix components integral for bone formation. RUNX2 physically interacts with a C-terminal domain in the long isoform of BRD4 and can co-occupy osteogenic enhancers. This BRD4 association is required for RUNX2 recruitment and appropriate osteoblast differentiation. We conclude that BRD4 controls facial bone development through osteoblast enhancer regulation of the RUNX2 transcriptional program.


Asunto(s)
Síndrome de Cornelia de Lange , Factores de Transcripción , Animales , Ratones , Proteínas de Ciclo Celular/genética , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Síndrome de Cornelia de Lange/genética , Cresta Neural/metabolismo , Proteínas Nucleares/metabolismo , Osteoblastos/metabolismo , Osteogénesis , Factores de Transcripción/metabolismo
7.
Small ; : e2311713, 2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38326098

RESUMEN

The discovery of non-precious catalysts for replacing the precious metal of ruthenium in the oxygen evolution reaction (OER) represents a key step in reducing the cost of green hydrogen production. The 2D d-MHOFs, a new 2D materials with controllable oxygen vacancies formed by controlling the degree of coordination bridging between metal hydroxyl oxide and BDC ligands are synthesized at room temperature, exhibit excellent OER properties with low overpotentials of 207  mV at 10 mA cm-2 . High-resolution transmission electron microscopy images and density functional theory calculations demonstrate that the introduction of oxygen vacancy sites leads to a lattice distortion and charge redistribution in the catalysts, enhancing the OER activity of 2D d-MHOFs comprehensively. Synchrotron radiation and in situ Raman/Fourier transform infrared spectroscopy indicate that part of oxygen defect sites on the surface of 2D d-MHOFs are prone to transition to highly active metal hydroxyl oxides during the OER process. This work provides a mild strategy for scalable preparation of 2D d-MHOFs nanosheets with controllable oxygen defects, reveals the relationship between oxygen vacancies and OER performance, and offers a profound insight into the basic process of structural transformation in the OER process.

8.
Genes Dev ; 30(5): 508-21, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26944678

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is a heterogeneous group of hematological tumors composed of distinct subtypes that vary in their genetic abnormalities, gene expression signatures, and prognoses. However, it remains unclear whether T-ALL subtypes differ at the functional level, and, as such, T-ALL treatments are uniformly applied across subtypes, leading to variable responses between patients. Here we reveal the existence of a subtype-specific epigenetic vulnerability in T-ALL by which a particular subgroup of T-ALL characterized by expression of the oncogenic transcription factor TAL1 is uniquely sensitive to variations in the dosage and activity of the histone 3 Lys27 (H3K27) demethylase UTX/KDM6A. Specifically, we identify UTX as a coactivator of TAL1 and show that it acts as a major regulator of the TAL1 leukemic gene expression program. Furthermore, we demonstrate that UTX, previously described as a tumor suppressor in T-ALL, is in fact a pro-oncogenic cofactor essential for leukemia maintenance in TAL1-positive (but not TAL1-negative) T-ALL. Exploiting this subtype-specific epigenetic vulnerability, we propose a novel therapeutic approach based on UTX inhibition through in vivo administration of an H3K27 demethylase inhibitor that efficiently kills TAL1-positive primary human leukemia. These findings provide the first opportunity to develop personalized epigenetic therapy for T-ALL patients.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica/genética , Terapia Genética , Histona Demetilasas/genética , Proteínas Nucleares/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Proteínas Proto-Oncogénicas/metabolismo , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Histona Demetilasas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/fisiopatología , Proteínas Proto-Oncogénicas/genética , Proteína 1 de la Leucemia Linfocítica T Aguda
9.
Genes Dev ; 30(2): 149-63, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26744420

RESUMEN

Class switch recombination (CSR) diversifies antibodies for productive immune responses while maintaining stability of the B-cell genome. Transcription at the immunoglobulin heavy chain (Igh) locus targets CSR-associated DNA damage and is promoted by the BRCT domain-containing PTIP (Pax transactivation domain-interacting protein). Although PTIP is a unique component of the mixed-lineage leukemia 3 (MLL3)/MLL4 chromatin-modifying complex, the mechanisms for how PTIP promotes transcription remain unclear. Here we dissected the minimal structural requirements of PTIP and its different protein complexes using quantitative proteomics in primary lymphocytes. We found that PTIP functions in transcription and CSR separately from its association with the MLL3/MLL4 complex and from its localization to sites of DNA damage. We identified a tandem BRCT domain of PTIP that is sufficient for CSR and identified PA1 as its main functional protein partner. Collectively, we provide genetic and biochemical evidence that a PTIP-PA1 subcomplex functions independently from the MLL3/MLL4 complex to mediate transcription during CSR. These results further our understanding of how multifunctional chromatin-modifying complexes are organized by subcomplexes that harbor unique and distinct activities.


Asunto(s)
Proteínas Portadoras/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Proteínas Nucleares/metabolismo , Daño del ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica/inmunología , Estructura Molecular , Estructura Terciaria de Proteína , Transporte de Proteínas
10.
J Am Chem Soc ; 145(44): 24425-24432, 2023 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-37880205

RESUMEN

Soft nanoporous crystals with structural dynamics are among the most exciting recently discovered materials. However, designing or controlling a porous system with delicate softness that can recognize similar gas pairs, particularly for the promoted ability at increased temperature, remains a challenge. Here, we report a soft crystal (NTU-68) with a one-dimensional (1D) channel that expands and contracts delicately around 4 Å at elevated temperature. The completely different adsorption processes of propane (C3H8: kinetic dominance) and propylene (C3H6: thermodynamic preference) allow the crystal to show a sieving separation of this mixtures (9.9 min·g-1) at 273 K, and the performance increases more than 2-fold (20.4 min·g-1) at 298 K. This phenomenon is contrary to the general observation for adsorption separation: the higher the temperature, the lower the efficiency. Gas-loaded in situ powder X-ray analysis and modeling calculations reveal that slight pore expansion caused by the increased temperature provides plausible nanochannel for adsorption of the relatively smaller C3H6 while maintaining constriction on the larger C3H8. In addition, the separation process remains unaffected by the general impurities, demonstrating its true potential as an alternative sorbent for practical applications. Moving forward, the delicate crystal dynamics and promoted capability for molecular recognition provide a new route for the design of next-generation sieve materials.

11.
Int J Cancer ; 153(3): 552-570, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37140208

RESUMEN

Although KMT2D, also known as MLL2, is known to play an essential role in development, differentiation, and tumor suppression, its role in pancreatic cancer development is not well understood. Here, we discovered a novel signaling axis mediated by KMT2D, which links TGF-ß to the activin A pathway. We found that TGF-ß upregulates a microRNA, miR-147b, which in turn leads to post-transcriptional silencing of KMT2D. Loss of KMT2D induces the expression and secretion of activin A, which activates a noncanonical p38 MAPK-mediated pathway to modulate cancer cell plasticity, promote a mesenchymal phenotype, and enhance tumor invasion and metastasis in mice. We observed a decreased KMT2D expression in human primary and metastatic pancreatic cancer. Furthermore, inhibition or knockdown of activin A reversed the protumoral role of KMT2D loss. These findings support a tumor-suppressive role of KMT2D in pancreatic cancer and identify miR-147b and activin A as novel therapeutic targets.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Plasticidad de la Célula , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Pancreáticas/patología , Factor de Crecimiento Transformador beta/metabolismo , Activinas/genética , Neoplasias Pancreáticas
12.
Development ; 147(21)2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32541010

RESUMEN

Kabuki syndrome (KS) is a congenital craniofacial disorder resulting from mutations in the KMT2D histone methylase (KS1) or the UTX histone demethylase (KS2). With small cohorts of KS2 patients, it is not clear whether differences exist in clinical manifestations relative to KS1. We mutated KMT2D in neural crest cells (NCCs) to study cellular and molecular functions in craniofacial development with respect to UTX. Similar to UTX, KMT2D NCC knockout mice demonstrate hypoplasia with reductions in frontonasal bone lengths. We have traced the onset of KMT2D and UTX mutant NCC frontal dysfunction to a stage of altered osteochondral progenitor differentiation. KMT2D NCC loss-of-function does exhibit unique phenotypes distinct from UTX mutation, including fully penetrant cleft palate, mandible hypoplasia and deficits in cranial base ossification. KMT2D mutant NCCs lead to defective secondary palatal shelf elevation with reduced expression of extracellular matrix components. KMT2D mutant chondrocytes in the cranial base fail to properly differentiate, leading to defective endochondral ossification. We conclude that KMT2D is required for appropriate cranial NCC differentiation and KMT2D-specific phenotypes may underlie differences between Kabuki syndrome subtypes.


Asunto(s)
Anomalías Múltiples/enzimología , Anomalías Múltiples/patología , Diferenciación Celular , Cara/anomalías , Enfermedades Hematológicas/enzimología , Enfermedades Hematológicas/patología , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Cresta Neural/enzimología , Cresta Neural/patología , Enfermedades Vestibulares/enzimología , Enfermedades Vestibulares/patología , Alelos , Animales , Linaje de la Célula , Movimiento Celular , Condrocitos/patología , Cara/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Mutación/genética , Osteogénesis , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Hueso Paladar/patología , Fenotipo , Cráneo/patología
13.
Inorg Chem ; 62(33): 13378-13386, 2023 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-37549317

RESUMEN

Zinc-air batteries (ZABs) have been considered as one of the most emerging systems for energy conversion and storage. However, the preparation of highly efficient oxygen reduction reaction (ORR) catalysts on an air cathode is still faced with significant challenges. Herein, we report a secondary nitrogen source strategy for fine-tuning the active center, which provides a carbon-based hierarchical porous catalyst (termed Co3O4@N/CNT-1000) for highly efficient ORR activity (E1/2 = 0.87 V, JL = 5.32 mA cm-2, and Eonset = 1.021 V) and excellent stability. Controlled experiments demonstrate that such high activity derives from the synergistic effect of cobalt tetroxide and bamboo-shaped carbon nanotubes doped with nitrogen, prepared by the pyrolysis of a two-dimensional metal-organic framework nanosheet (termed NTU-70) and melamine. Furthermore, the ZAB assembled with Co3O4@N/CNT-1000 displays high specific capacity (854 mA h g-1Zn) and power density (179 mW cm-2), excellent long-term cycling (330 h), and durable charging/discharging ability.

14.
Inorg Chem ; 62(21): 8219-8231, 2023 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-37191263

RESUMEN

An effective approach to synthesize polycrystalline Ni-Co-Mo sulfide (NiCoMoS) is developed through doping engineering coupled with chemical transformation. The polycrystalline NiCoMoS with enriched active edge sites is designed and fabricated on a Ni foam (NF) via a facile hydrothermal calcination and post-sulfidation approach, where the polycrystalline NiCoMoO4 precursor is elaborately prepared by doping Co ions into the NiMoO4 lattice and subsequently in-situ-converted into NiCoMoS with 3D architectures of ordered nanoneedle arrays. Benefiting from the unique 3D structure and synergistic effects of each component, the optimized needle-like NiCoMoS(2.0) arraying on a NF as a self-standing electrode exhibits superior electrochemical performances with a high specific charge (920.0 C g-1 at 1.0 A g-1), excellent rate capability, and good long-term stability. Furthermore, the assembled NiCoMoS//activated carbon hybrid device presents a satisfactory supercapacitor performance, affording an energy density of 35.2 W h kg-1 at a power density of 800.0 W kg-1 and competitive long-term stability (83.8% retention at 15 A g-1 after 10,000 cycles). Such a novel strategy may pave a new route for exploring other polymetallic sulfides with enriched, exposed active edge sites for energy-related applications.

15.
Nature ; 535(7612): 382-7, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27443740

RESUMEN

Cells deficient in the Brca1 and Brca2 genes have reduced capacity to repair DNA double-strand breaks by homologous recombination and consequently are hypersensitive to DNA-damaging agents, including cisplatin and poly(ADP-ribose) polymerase (PARP) inhibitors. Here we show that loss of the MLL3/4 complex protein, PTIP, protects Brca1/2-deficient cells from DNA damage and rescues the lethality of Brca2-deficient embryonic stem cells. However, PTIP deficiency does not restore homologous recombination activity at double-strand breaks. Instead, its absence inhibits the recruitment of the MRE11 nuclease to stalled replication forks, which in turn protects nascent DNA strands from extensive degradation. More generally, acquisition of PARP inhibitors and cisplatin resistance is associated with replication fork protection in Brca2-deficient tumour cells that do not develop Brca2 reversion mutations. Disruption of multiple proteins, including PARP1 and CHD4, leads to the same end point of replication fork protection, highlighting the complexities by which tumour cells evade chemotherapeutic interventions and acquire drug resistance.


Asunto(s)
Replicación del ADN/fisiología , Resistencia a Antineoplásicos/efectos de los fármacos , Eliminación de Gen , Genes BRCA1 , Genes BRCA2 , Neoplasias/patología , Proteínas Nucleares/deficiencia , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Cisplatino/farmacología , ADN/biosíntesis , ADN/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , ADN Helicasas/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos/genética , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Femenino , Recombinación Homóloga , Proteína Homóloga de MRE11 , Ratones , Neoplasias/genética , Proteínas Nucleares/genética , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/genética
16.
Nucleic Acids Res ; 48(10): 5442-5456, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32365172

RESUMEN

In mammalian cells, distinct H3K4 methylation states are created by deposition of methyl groups by multiple complexes of histone lysine methyltransferase 2 (KMT2) family proteins. For comprehensive analyses that directly compare the catalytic properties of all six human KMT2 complexes, we employed a biochemically defined system reconstituted with recombinant KMT2 core complexes (KMT2CoreCs) containing minimal components required for nucleosomal H3K4 methylation activity. We found that each KMT2CoreC generates distinct states and different levels of H3K4 methylation, and except for MLL3 all are stimulated by H2Bub. Notably, SET1BCoreC exhibited the strongest H3K4 methylation activity and, to our surprise, did not require H2B ubiquitylation (H2Bub); in contrast, H2Bub was required for the H3K4me2/3 activity of the paralog SET1ACoreC. We also found that WDR5, RbBP5, ASH2L and DPY30 are required for efficient H3K4 methyltransferase activities of all KMT2CoreCs except MLL3, which could produce H3K4me1 in the absence of WDR5. Importantly, deletion of the PHD2 domain of CFP1 led to complete loss of the H3K4me2/3 activities of SET1A/BCoreCs in the presence of H2Bub, indicating a critical role for this domain in the H2Bub-stimulated H3K4 methylation. Collectively, our results suggest that each KMT2 complex methylates H3K4 through distinct mechanisms in which individual subunits differentially participate.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Ubiquitinación , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/química , Humanos , Metilación , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Nucleosomas/enzimología , Dominios Proteicos , Subunidades de Proteína/metabolismo
17.
Zhongguo Zhong Yao Za Zhi ; 47(5): 1359-1369, 2022 Mar.
Artículo en Zh | MEDLINE | ID: mdl-35343165

RESUMEN

UPLC-Q-TOF-MS combined with network pharmacology and experimental verification was used to explore the mechanism of acupoint sticking therapy(AST) in the intervention of bronchial asthma(BA). The chemical components of Sinapis Semen, Cory-dalis Rhizoma, Kansui Radix, Asari Radix et Rhizoma, and Zingiberis Rhizoma Recens were retrieved from TCMSP as self-built database. The active components in AST drugs were analyzed by UPLC-Q-TOF-MS, and the targets were screened out in TCMSP and Swiss-TargetPrediction. Targets of BA were collected from GeneCards, and the intersection of active components and targets was obtained by Venny 2.1.0. The potential targets were imported into STRING and DAVID for PPI, GO, and KEGG analyses. The asthma model induced by house dust mite(HDM) was established in mice. The mechanism of AST on asthmatic mice was explored by pulmonary function, Western blot, and flow cytometry. The results indicated that 54 active components were obtained by UPLC-Q-TOF-MS and 162 potential targets were obtained from the intersection. The first 53 targets were selected as key targets. PPI, GO, and KEGG analyses showed that AST presumedly acted on SRC, PIK3 CA, and other targets through active components such as sinoacutine, sinapic acid, dihydrocapsaicin, and 6-gingerol and regulated PI3 K-AKT, ErbB, chemokine, sphingolipid, and other signaling pathways to intervene in the pathological mechanism of BA. AST can improve lung function, down-regulate the expression of PI3 K and p-AKT proteins in lung tissues, enhance the expression of PETN protein, and reduce the level of type Ⅱ innate immune cells(ILC2 s) in lung tissues of asthmatic mice. In conclusion, AST may inhibit ILC2 s by down-regulating the PI3 K-AKT pathway to relieve asthmatic airway inflammation and reduce airway hyperresponsiveness.


Asunto(s)
Puntos de Acupuntura , Asma , Animales , Asma/tratamiento farmacológico , Medicamentos Herbarios Chinos , Inmunidad Innata , Linfocitos , Ratones , Farmacología en Red
18.
J Biol Chem ; 295(49): 16877-16887, 2020 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-32994220

RESUMEN

Histone recognition by "reader" modules serves as a fundamental mechanism in epigenetic regulation. Previous studies have shown that Spindlin1 is a reader of histone H3K4me3 as well as "K4me3-R8me2a" and promotes transcription of rDNA or Wnt/TCF4 target genes. Here we show that Spindlin1 also acts as a potent reader of histone H3 "K4me3-K9me3/2" bivalent methylation pattern. Calorimetric titration revealed a binding affinity of 16 nm between Spindlin1 and H3 "K4me3-K9me3" peptide, which is one to three orders of magnitude stronger than most other histone readout events at peptide level. Structural studies revealed concurrent recognition of H3K4me3 and H3K9me3/2 by aromatic pockets 2 and 1 of Spindlin1, respectively. Epigenomic profiling studies showed that Spindlin1 colocalizes with both H3K4me3 and H3K9me3 peaks in a subset of genes enriched in biological processes of transcription and its regulation. Moreover, the distribution of Spindlin1 peaks is primarily associated with H3K4me3 but not H3K9me3, which suggests that Spindlin1 is a downstream effector of H3K4me3 generated in heterochromatic regions. Collectively, our work calls attention to an intriguing function of Spindlin1 as a potent H3 "K4me3-K9me3/2" bivalent mark reader, thereby balancing gene expression and silencing in H3K9me3/2-enriched regions.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Histonas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Calorimetría , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cristalografía por Rayos X , Bases de Datos de Proteínas , Epigenómica , Expresión Génica , Histonas/química , Histonas/genética , Humanos , Enlace de Hidrógeno , Metilación , Proteínas Asociadas a Microtúbulos/química , Proteínas Asociadas a Microtúbulos/genética , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Fosfoproteínas/química , Fosfoproteínas/genética , Unión Proteica , Estructura Cuaternaria de Proteína
19.
Small ; 17(40): e2102425, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34494368

RESUMEN

Designing highly efficient and durable electrocatalysts that accelerate sluggish oxygen reduction reaction kinetics for fuel cells and metal-air batteries are highly desirable but challenging. Herein, a facile yet robust strategy is reported to rationally design single iron active centers synergized with local S atoms in metal-organic frameworks derived from hierarchically porous carbon nanorods (Fe/N,S-HC). The cooperative trithiocyanuric acid-based coating not only introduces S atoms that regulate the coordination environment of the active centers, but also facilitates the formation of a hierarchically porous structure. Benefiting from electronic modulation and architectural functionality, Fe/N,S-HC catalyst shows markedly enhanced ORR performance with a half-wave potential (E1/2 ) of 0.912 V and satisfactory long-term durability in alkaline medium, outperforming those of commercial Pt/C. Impressively, Fe/N,S-HC-based Zn-air battery also presents outstanding battery performance and long-term stability. Both electrochemical experimental and density functional theoretical (DFT) calculated results suggest that the FeN4 sites tailored with local S atoms are favorable for the adsorption/desorption of oxygen intermediate, resulting in lower activation energy barrier and ultraefficient oxygen reduction catalytic activity. This work provides an atomic-level combined with porous morphological-level insights into oxygen reduction catalytic property, promoting rational design and development of novel highly efficient single-atom catalysts for the renewable energy applications.

20.
Immunity ; 37(6): 971-85, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23159437

RESUMEN

Histone 3 lysine 4 trimethylation (H3K4me3) is associated with promoters of active genes and found at hot spots for DNA recombination. Here we have shown that PAXIP1 (also known as PTIP), a protein associated with MLL3 and MLL4 methyltransferase and the DNA damage response, regulates RAG-mediated cleavage and repair during V(D)J recombination in CD4(+) CD8(+) DP thymocytes. Loss of PAXIP1 in developing thymocytes diminished Jα H3K4me3 and germline transcription, suppressed double strand break formation at 3' Jα segments, but resulted in accumulation of unresolved T cell receptor α-chain gene (Tcra) breaks. Moreover, PAXIP1 was essential for release of mature single positive (SP) αß T cells from the thymus through transcriptional activation of sphingosine-1-phosphate receptor S1pr1 as well as for natural killer T cell development. Thus, in addition to maintaining genome integrity during Tcra rearrangements, PAXIP1 controls distinct transcriptional programs during DP differentiation necessary for Tcra locus accessibility, licensing mature thymocytes for trafficking and natural killer T cell development.


Asunto(s)
Proteínas Portadoras/genética , Diferenciación Celular , Movimiento Celular , Daño del ADN , Regulación de la Expresión Génica , Proteínas Nucleares/genética , Timocitos/citología , Timocitos/inmunología , Animales , Proteínas Portadoras/metabolismo , Linaje de la Célula/genética , Movimiento Celular/genética , Proteínas de Unión al ADN , Histonas/metabolismo , Ratones , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/metabolismo , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Lisoesfingolípidos/genética , Recombinación Genética , Receptores de Esfingosina-1-Fosfato , Linfocitos T/citología , Linfocitos T/metabolismo , Timocitos/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA