Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38766219

RESUMEN

Cell differentiation during organogenesis relies on precise epigenetic and transcriptional control. Disruptions to this regulation can result in developmental abnormalities and malignancies, yet the underlying mechanisms are not well understood. Wilms tumors, a type of embryonal tumor closely linked to disrupted organogenesis, harbor mutations in epigenetic regulators in 30-50% of cases. However, the role of these regulators in kidney development and pathogenesis remains unexplored. By integrating mouse modeling, histological characterizations, and single-cell transcriptomics and chromatin accessibility profiling, we show that a Wilms tumor-associated mutation in the chromatin reader protein ENL disrupts kidney development trajectory by rewiring the gene regulatory landscape. Specifically, the mutant ENL promotes the commitment of nephron progenitors while simultaneously restricting their differentiation by dysregulating key transcription factor regulons, particularly the HOX clusters. It also induces the emergence of abnormal progenitor cells that lose their chromatin identity associated with kidney specification. Furthermore, the mutant ENL might modulate stroma-nephron interactions via paracrine Wnt signaling. These multifaceted effects caused by the mutation result in severe developmental defects in the kidney and early postnatal mortality in mice. Notably, transient inhibition of the histone acetylation binding activity of mutant ENL with a small molecule displaces transcriptional condensates formed by mutant ENL from target genes, abolishes its gene activation function, and restores developmental defects in mice. This work provides new insights into how mutations in epigenetic regulators can alter the gene regulatory landscape to disrupt kidney developmental programs at single-cell resolution in vivo . It also offers a proof-of-concept for the use of epigenetics-targeted agents to rectify developmental defects.

2.
Curr Opin Genet Dev ; 86: 102203, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38788489

RESUMEN

Precise gene expression, crucial for normal development and health, depends on the co-ordinated assembly and function of various factors within the crowded nucleus. Recent evidence suggests that this process is in part regulated by mesoscale compartmentalization and concentration of transcriptional components within condensates, offering a new perspective on gene regulation. Dysregulation of transcriptional condensates is increasingly associated with diseases, indicating a potential role in pathogenesis. In this mini-review, we provide a concise overview of the current understanding of the formation and function of transcriptional condensates, with a specific focus on recent advances in their dysregulation and implications in diseases, notably cancer. We also address limitations in the field and highlight open questions for future research.


Asunto(s)
Regulación de la Expresión Génica , Neoplasias , Transcripción Genética , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Regulación de la Expresión Génica/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Cancer Discov ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38655899

RESUMEN

Gain-of-function mutations in the histone acetylation 'reader' ENL, found in AML and Wilms tumor, are known to drive condensate formation and gene activation in cellular systems. However, their role in tumorigenesis remains unclear. Using a conditional knock-in mouse model, we show that mutant ENL perturbs normal hematopoiesis, induces aberrant expansion of myeloid progenitors, and triggers rapid onset of aggressive AML. Mutant ENL alters developmental and inflammatory gene programs in part by remodeling histone modifications. Mutant ENL forms condensates in hematopoietic stem/progenitor cells at key leukemogenic genes, and disrupting condensate formation via mutagenesis impairs its chromatin and oncogenic function. Moreover, treatment with an acetyl-binding inhibitor of mutant ENL displaces these condensates from target loci, inhibits mutant ENL-induced chromatin changes, and delays AML initiation and progression in vivo. Our study elucidates the function of ENL mutations in chromatin regulation and tumorigenesis, and demonstrates the potential of targeting pathogenic condensates in cancer treatment.

4.
ACS Med Chem Lett ; 15(4): 524-532, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38628784

RESUMEN

Eleven-nineteen leukemia (ENL) is an epigenetic reader protein that drives oncogenic transcriptional programs in acute myeloid leukemia (AML). AML is one of the deadliest hematopoietic malignancies, with an overall 5-year survival rate of 27%. The epigenetic reader activity of ENL is mediated by its YEATS domain that binds to acetyl and crotonyl marks on histone tails and colocalizes with promoters of actively transcribed genes that are essential for leukemia. Prior to the discovery of TDI-11055, existing inhibitors of ENL YEATS showed in vitro potency, but had not shown efficacy in in vivo animal models. During the course of the medicinal chemistry campaign described here, we identified ENL YEATS inhibitor TDI-11055 that has an improved pharmacokinetic profile and is appropriate for in vivo evaluation of the ENL YEATS inhibition mechanism in AML.

5.
Nat Commun ; 14(1): 4403, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37479684

RESUMEN

The p53 tumor suppressor regulates multiple context-dependent tumor suppressive programs. Although p53 is mutated in ~90% of small cell lung cancer (SCLC) tumors, how p53 mediates tumor suppression in this context is unknown. Here, using a mouse model of SCLC in which endogenous p53 expression can be conditionally and temporally regulated, we show that SCLC tumors maintain a requirement for p53 inactivation. However, we identify tumor subtype heterogeneity between SCLC tumors such that p53 reactivation induces senescence in a subset of tumors, while in others, p53 induces necrosis. We pinpoint cyclophilins as critical determinants of a p53-induced transcriptional program that is specific to SCLC tumors and cell lines poised to undergo p53-mediated necrosis. Importantly, inhibition of cyclophilin isomerase activity, or genetic ablation of specific cyclophilin genes, suppresses p53-mediated necrosis by limiting p53 transcriptional output without impacting p53 chromatin binding. Our study demonstrates that intertumoral heterogeneity in SCLC influences the biological response to p53 restoration, describes a cyclophilin-dependent mechanism of p53-regulated cell death, and uncovers putative mechanisms for the treatment of this most-recalcitrant tumor type.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Ciclofilinas/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Proteína p53 Supresora de Tumor/genética , Necrosis/genética , Neoplasias Pulmonares/genética
6.
Blood ; 142(11): 973-988, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37235754

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma. Up to 40% of patients with DLBCL display refractory disease or relapse after standard chemotherapy treatment (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone [R-CHOP]), leading to significant morbidity and mortality. The molecular mechanisms of chemoresistance in DLBCL remain incompletely understood. Using a cullin-really interesting new gene (RING) ligase-based CRISPR-Cas9 library, we identify that inactivation of the E3 ubiquitin ligase KLHL6 promotes DLBCL chemoresistance. Furthermore, proteomic approaches helped identify KLHL6 as a novel master regulator of plasma membrane-associated NOTCH2 via proteasome-dependent degradation. In CHOP-resistant DLBCL tumors, mutations of NOTCH2 result in a protein that escapes the mechanism of ubiquitin-dependent proteolysis, leading to protein stabilization and activation of the oncogenic RAS signaling pathway. Targeting CHOP-resistant DLBCL tumors with the phase 3 clinical trial molecules nirogacestat, a selective γ-secretase inhibitor, and ipatasertib, a pan-AKT inhibitor, synergistically promotes DLBCL destruction. These findings establish the rationale for therapeutic strategies aimed at targeting the oncogenic pathway activated in KLHL6- or NOTCH2-mutated DLBCL.


Asunto(s)
Resistencia a Antineoplásicos , Linfoma de Células B Grandes Difuso , Humanos , Resistencia a Antineoplásicos/genética , Ubiquitina , Proteómica , Recurrencia Local de Neoplasia/tratamiento farmacológico , Rituximab/uso terapéutico , Vincristina , Ciclofosfamida , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Prednisona , Mutación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Receptor Notch2/genética
8.
Cancer Discov ; 13(3): 724-745, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36455589

RESUMEN

Nucleophosmin (NPM1) is a ubiquitously expressed nucleolar protein with a wide range of biological functions. In 30% of acute myeloid leukemia (AML), the terminal exon of NPM1 is often found mutated, resulting in the addition of a nuclear export signal and a shift of the protein to the cytoplasm (NPM1c). AMLs carrying this mutation have aberrant expression of the HOXA/B genes, whose overexpression leads to leukemogenic transformation. Here, for the first time, we comprehensively prove that NPM1c binds to a subset of active gene promoters in NPM1c AMLs, including well-known leukemia-driving genes-HOXA/B cluster genes and MEIS1. NPM1c sustains the active transcription of key target genes by orchestrating a transcription hub and maintains the active chromatin landscape by inhibiting the activity of histone deacetylases. Together, these findings reveal the neomorphic function of NPM1c as a transcriptional amplifier for leukemic gene expression and open up new paradigms for therapeutic intervention. SIGNIFICANCE: NPM1 mutation is the most common mutation in AML, yet the mechanism of how the mutant protein results in AML remains unclear. Here, for the first time, we prove mutant NPM1 directly binds to active chromatin regions and hijacks the transcription of AML-driving genes. See related article by Uckelmann et al., p. 746. This article is highlighted in the In This Issue feature, p. 517.


Asunto(s)
Leucemia Mieloide Aguda , Nucleofosmina , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Mutación , Cromatina/genética
9.
Mol Cell ; 82(21): 4080-4098.e12, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36272410

RESUMEN

Growing evidence suggests prevalence of transcriptional condensates on chromatin, yet their mechanisms of formation and functional significance remain largely unclear. In human cancer, a series of mutations in the histone acetylation reader ENL create gain-of-function mutants with increased transcriptional activation ability. Here, we show that these mutations, clustered in ENL's structured acetyl-reading YEATS domain, trigger aberrant condensates at native genomic targets through multivalent homotypic and heterotypic interactions. Mechanistically, mutation-induced structural changes in the YEATS domain, ENL's two disordered regions of opposing charges, and the incorporation of extrinsic elongation factors are all required for ENL condensate formation. Extensive mutagenesis establishes condensate formation as a driver of oncogenic gene activation. Furthermore, expression of ENL mutants beyond the endogenous level leads to non-functional condensates. Our findings provide new mechanistic and functional insights into cancer-associated condensates and support condensate dysregulation as an oncogenic mechanism.


Asunto(s)
Neoplasias , Cuerpos Nucleares , Humanos , Dominios Proteicos , Cromatina/genética , Mutación , Neoplasias/genética
10.
Cancer Discov ; 12(11): 2684-2709, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36053276

RESUMEN

The chromatin reader eleven-nineteen leukemia (ENL) has been identified as a critical dependency in acute myeloid leukemia (AML), but its therapeutic potential remains unclear. We describe a potent and orally bioavailable small-molecule inhibitor of ENL, TDI-11055, which displaces ENL from chromatin by blocking its YEATS domain interaction with acylated histones. Cell lines and primary patient samples carrying MLL rearrangements or NPM1 mutations are responsive to TDI-11055. A CRISPR-Cas9-mediated mutagenesis screen uncovers an ENL mutation that confers resistance to TDI-11055, validating the compound's on-target activity. TDI-11055 treatment rapidly decreases chromatin occupancy of ENL-associated complexes and impairs transcription elongation, leading to suppression of key oncogenic gene expression programs and induction of differentiation. In vivo treatment with TDI-11055 blocks disease progression in cell line- and patient-derived xenograft models of MLL-rearranged and NPM1-mutated AML. Our results establish ENL displacement from chromatin as a promising epigenetic therapy for molecularly defined AML subsets and support the clinical translation of this approach. SIGNIFICANCE: AML is a poor-prognosis disease for which new therapeutic approaches are desperately needed. We developed an orally bioavailable inhibitor of ENL, demonstrated its potent efficacy in MLL-rearranged and NPM1-mutated AML, and determined its mechanisms of action. These biological and chemical insights will facilitate both basic research and clinical translation. This article is highlighted in the In This Issue feature, p. 2483.


Asunto(s)
Leucemia Mieloide Aguda , Lisina , Humanos , Leucemia Mieloide Aguda/genética , Histonas/metabolismo , Cromatina , Proteína de la Leucemia Mieloide-Linfoide/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-35356238

RESUMEN

Background: Re Du Ning, a traditional Chinese medicine injection, has been widely used for the treatment of chronic obstructive pulmonary disease, although without established systematic review evidence. This systematic review aimed to assess the efficacy and safety of Re Du Ning in the treatment of acute exacerbations of chronic obstructive pulmonary disease (AECOPD). Methods: We searched seven databases (PubMed, Embase, the Cochrane Library, SinoMed, CNKI, WanFang, and the Chinese Clinical Trial Registry) up to November 1, 2021, to identify randomized controlled trials of Re Du Ning for AECOPD. Two researchers independently carried out literature screening and data extraction. Effects were measured by risk ratios (RRs) or mean differences (MDs) with 95% confidence intervals (CIs). The meta-analysis was completed by RevMan 5.4 software. Results: Twenty-six studies met the eligibility criteria, with a total of 2284 patients. The findings of the meta-analysis indicated that the response rate of the experimental group was higher than that of the control group: RR = 1.14% and 95% CI: (1.09, 1.19). Significantly greater improvements in pulmonary function: FEV1: MD = 0.28 L, 95% CI: (0.20, 0.36); FEV1/FVC: MD = 8.63%, 95% CI: (4.68, 12.59); T-lymphocyte counts: CD4: MD = 6%, 95% CI: (2.44, 9.56); CD3: MD = 10.42%, 95% CI: (8.6, 12.24); CD4/CD8: MD = 0.38%, 95% CI: (0.32, 0.43); acid/base imbalance: PH: MD = 0.05, 95% CI: (0.01, 0.10); PaO2: MD = 9.02 mmHg, 95% CI: (11.11, 0.10), p=0.005; C-reactive protein: MD = -6.65 mg/L, 95% CI: (-10.97, -2.34); and PCT: MD = -0.28 µg/L, 95% (CI: -0.41, -0.15) were observed in patients receiving Re Du Ning compared with those receiving the control treatment. Re Du Ning did not significantly change the carbon dioxide partial pressure. All reported adverse reactions were mild. Conclusion: Re Du Ning injection, as a complementary therapy to routine treatment, has better efficacy than Western medicine alone in relieving clinical symptoms, delaying pulmonary function decline, and improving inflammation indicators for AECOPD, with good safety. The evidence was limited by a lack of high-quality RCTs.

12.
Nat Cancer ; 3(1): 60-74, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121988

RESUMEN

Despite increased overall survival rates, curative options for metastatic breast cancer remain limited. We have previously shown that metadherin (MTDH) is frequently overexpressed in poor prognosis breast cancer, where it promotes metastasis and therapy resistance through its interaction with staphylococcal nuclease domain-containing 1 (SND1). Through genetic and pharmacological targeting of the MTDH-SND1 interaction, we reveal a key role for this complex in suppressing antitumor T cell responses in breast cancer. The MTDH-SND1 complex reduces tumor antigen presentation and inhibits T cell infiltration and activation by binding to and destabilizing Tap1/2 messenger RNAs, which encode key components of the antigen-presentation machinery. Following small-molecule compound C26-A6 treatment to disrupt the MTDH-SND1 complex, we showed enhanced immune surveillance and sensitivity to anti-programmed cell death protein 1 therapy in preclinical models of metastatic breast cancer, in support of this combination therapy as a viable approach to increase immune-checkpoint blockade therapy responses in metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama , Presentación de Antígeno , Neoplasias de la Mama/tratamiento farmacológico , Endonucleasas/metabolismo , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Nucleasa Microcócica/metabolismo , Proteínas Nucleares/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
13.
Nat Cancer ; 3(1): 43-59, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35121987

RESUMEN

Metastatic breast cancer is a leading health burden worldwide. Previous studies have shown that metadherin (MTDH) promotes breast cancer initiation, metastasis and therapy resistance; however, the therapeutic potential of targeting MTDH remains largely unexplored. Here, we used genetically modified mice and demonstrate that genetic ablation of Mtdh inhibits breast cancer development through disrupting the interaction with staphylococcal nuclease domain-containing 1 (SND1), which is required to sustain breast cancer progression in established tumors. We performed a small-molecule compound screening to identify a class of specific inhibitors that disrupts the protein-protein interaction (PPI) between MTDH and SND1 and show that our lead candidate compounds C26-A2 and C26-A6 suppressed tumor growth and metastasis and enhanced chemotherapy sensitivity in preclinical models of triple-negative breast cancer (TNBC). Our results demonstrate a significant therapeutic potential in targeting the MTDH-SND1 complex and identify a new class of therapeutic agents for metastatic breast cancer.


Asunto(s)
Endonucleasas/metabolismo , Proteínas de la Membrana/metabolismo , Nucleasa Microcócica , Proteínas de Unión al ARN/metabolismo , Neoplasias de la Mama Triple Negativas , Animales , Moléculas de Adhesión Celular/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Proteínas de Unión al ARN/genética , Factores de Transcripción
14.
Cancer Discov ; 12(3): 792-811, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34853079

RESUMEN

Epigenetic programs are dysregulated in acute myeloid leukemia (AML) and help enforce an oncogenic state of differentiation arrest. To identify key epigenetic regulators of AML cell fate, we performed a differentiation-focused CRISPR screen in AML cells. This screen identified the histone acetyltransferase KAT6A as a novel regulator of myeloid differentiation that drives critical leukemogenic gene-expression programs. We show that KAT6A is the initiator of a newly described transcriptional control module in which KAT6A-catalyzed promoter H3K9ac is bound by the acetyl-lysine reader ENL, which in turn cooperates with a network of chromatin factors to induce transcriptional elongation. Inhibition of KAT6A has strong anti-AML phenotypes in vitro and in vivo, suggesting that KAT6A small-molecule inhibitors could be of high therapeutic interest for mono-therapy or combinatorial differentiation-based treatment of AML. SIGNIFICANCE: AML is a poor-prognosis disease characterized by differentiation blockade. Through a cell-fate CRISPR screen, we identified KAT6A as a novel regulator of AML cell differentiation. Mechanistically, KAT6A cooperates with ENL in a "writer-reader" epigenetic transcriptional control module. These results uncover a new epigenetic dependency and therapeutic opportunity in AML. This article is highlighted in the In This Issue feature, p. 587.


Asunto(s)
Leucemia Mieloide Aguda , Oncogenes , Cromatina/genética , Epigénesis Genética , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Proteínas de Neoplasias , Proteínas Nucleares , Factores de Transcripción
16.
Cancer Res ; 81(4): 1014-1025, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33239430

RESUMEN

Colorectal and lung cancers account for one-third of all cancer-related deaths worldwide. Previous studies suggested that metadherin (MTDH) is involved in the development of colorectal and lung cancers. However, how MTDH regulates the pathogenesis of these cancers remains largely unknown. Using genetically modified mouse models of spontaneous colorectal and lung cancers, we found that MTDH promotes cancer progression by facilitating Wnt activation and by inducing cytotoxic T-cell exhaustion, respectively. Moreover, we developed locked nucleic acid-modified (LNA) MTDH antisense oligonucleotides (ASO) that effectively and specifically suppress MTDH expression in vitro and in vivo. Treatments with MTDH ASOs in mouse models significantly attenuated progression and metastasis of colorectal, lung, and breast cancers. Our study opens a new avenue for developing therapies against colorectal and lung cancers by targeting MTDH using LNA-modified ASO. SIGNIFICANCE: This study provides new insights into the mechanism of MTDH in promoting colorectal and lung cancers, as well as genetic and pharmacologic evidence supporting the development of MTDH-targeting therapeutics.


Asunto(s)
Adenocarcinoma/terapia , Neoplasias Colorrectales/terapia , Neoplasias Pulmonares/terapia , Proteínas de la Membrana/antagonistas & inhibidores , Oligonucleótidos Antisentido/uso terapéutico , Proteínas de Unión al ARN/antagonistas & inhibidores , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Terapia Genética/métodos , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Metástasis de la Neoplasia , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Proteínas de Unión al ARN/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Nature ; 577(7788): 121-126, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31853060

RESUMEN

Modifications of histone proteins have essential roles in normal development and human disease. Recognition of modified histones by 'reader' proteins is a key mechanism that mediates the function of histone modifications, but how the dysregulation of these readers might contribute to disease remains poorly understood. We previously identified the ENL protein as a reader of histone acetylation via its YEATS domain, linking it to the expression of cancer-driving genes in acute leukaemia1. Recurrent hotspot mutations have been found in the ENL YEATS domain in Wilms tumour2,3, the most common type of paediatric kidney cancer. Here we show, using human and mouse cells, that these mutations impair cell-fate regulation by conferring gain-of-function in chromatin recruitment and transcriptional control. ENL mutants induce gene-expression changes that favour a premalignant cell fate, and, in an assay for nephrogenesis using murine cells, result in undifferentiated structures resembling those observed in human Wilms tumour. Mechanistically, although bound to largely similar genomic loci as the wild-type protein, ENL mutants exhibit increased occupancy at a subset of targets, leading to a marked increase in the recruitment and activity of transcription elongation machinery that enforces active transcription from target loci. Furthermore, ectopically expressed ENL mutants exhibit greater self-association and form discrete and dynamic nuclear puncta that are characteristic of biomolecular hubs consisting of local high concentrations of regulatory factors. Such mutation-driven ENL self-association is functionally linked to enhanced chromatin occupancy and gene activation. Collectively, our findings show that hotspot mutations in a chromatin-reader domain drive self-reinforced recruitment, derailing normal cell-fate control during development and leading to an oncogenic outcome.


Asunto(s)
Linaje de la Célula , Cromatina/genética , Proteínas de Unión al ADN/metabolismo , Mutación con Ganancia de Función , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Ratones , Nefronas/metabolismo , Nefronas/patología , Factores de Transcripción/química , Factores de Transcripción/genética
18.
Nat Chem Biol ; 14(12): 1140-1149, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30374167

RESUMEN

Chemical probes of epigenetic 'readers' of histone post-translational modifications (PTMs) have become powerful tools for mechanistic and functional studies of their target proteins in normal physiology and disease pathogenesis. Here we report the development of the first class of chemical probes of YEATS domains, newly identified 'readers' of histone lysine acetylation (Kac) and crotonylation (Kcr). Guided by the structural analysis of a YEATS-Kcr complex, we developed a series of peptide-based inhibitors of YEATS domains by targeting a unique π-π-π stacking interaction at the proteins' Kcr recognition site. Further structure optimization resulted in the selective inhibitors preferentially binding to individual YEATS-containing proteins including AF9 and ENL with submicromolar affinities. We demonstrate that one of the ENL YEATS-selective inhibitors, XL-13m, engages with endogenous ENL, perturbs the recruitment of ENL onto chromatin, and synergizes the BET and DOT1L inhibition-induced downregulation of oncogenes in MLL-rearranged acute leukemia.


Asunto(s)
Diseño de Fármacos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Péptidos/farmacología , Factores de Elongación Transcripcional/antagonistas & inhibidores , Azepinas/farmacología , Línea Celular , Cromatina/metabolismo , Cristalografía por Rayos X , Regulación de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina , Humanos , Lisina/metabolismo , Metiltransferasas/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Péptidos/química , Dominios Proteicos , Procesamiento Proteico-Postraduccional , Relación Estructura-Actividad , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/metabolismo , Triazoles/farmacología
19.
Zhongguo Zhen Jiu ; 37(8): 869-873, 2017 Aug 12.
Artículo en Chino | MEDLINE | ID: mdl-29231349

RESUMEN

OBJECTIVE: To observe the clinical effect of ear point embedding on plasma and effect site concentrations of propofol-remifentanil in elderly patients who underwent abdominal external hernia surgery at the time of consciousness and pain disappearing by target-controlled infusion (TCI) and bispectral index (BIS). METHODS: Fifty patients who underwent elective abdominal hernia surgery were randomly assigned into an observation group and a control group, 25 cases in each one. In the observation group, 30 minutes before anesthesia induction, Fugugou (Extra), Gan (CO12), Pizhixia (AT4), and Shenmen (TF4) were embedded by auricular needles until the end of surgery, 10 times of counter press each point. In the control group, the same amount of auricular tape was applied until the end of surgery at the same points without stimulation 30 minutes before anesthesia induction. Patients in the two groups were given total intravenous anesthesia, and BIS was monitored by BIS anesthesia depth monitor. Propofol was infused by TCI at a beginning concentration of 1.5µg/L and increased by 0.3µg/L every 30s until the patients lost their consciousness. After that, remifentanil was infused by TCI at a beginning concentration of 2.0µg/L and increased by 0.3µg/L every 30s until the patients had no body reaction to pain stimulation (orbital reflex). Indices were recorded, including mean arterial pressure (MAP), heart rate (HR) and the BIS values, at the time of T0 (entering into the operation room), T1 (losing consciousness) and T2 (pain relief), the plasma and effect site concentrations of propofol at T1, the plasma and effect site concentrations of remifentanil at T2. After surgery we recorded the total amounts of propofol and remifentanil, surgery time and anesthesia time. RESULTS: At T1 and T2, MAP and HR of the observation group were higher than those of the control group (P<0.05, P<0.01). At T1, the plasma and effect site concentrations of propofol in the observation group were significantly lower than those in the control group (P<0.05, P<0.01). At T2, the plasma and effect site concentrations of remifentanil in the observation group were significantly lower than those in the control group (P<0.05, P<0.01). There was no significant difference in BIS values at T1 and T2 between the two groups (bothP>0.05). There was no significant difference in operation time and anesthesia time between the two groups (bothP>0.05). The total amount of remifentanil in the observation group was significantly lower than that in the control group (P<0.01). There was no significant difference in the total amount of propofol between the two groups (P>0.05). CONCLUSIONS: Ear points embedding combined with propofol-remifentanil TCI could reduce the plasma and effect site concentrations of propofol and remifentanil and the total amount of remifentanil in elderly patients with extra-abdominal hernia surgery, and had the effect of assisting sedation and analgesia.


Asunto(s)
Analgesia por Acupuntura/métodos , Acupuntura Auricular/métodos , Anestesia Intravenosa/métodos , Anestésicos Intravenosos/administración & dosificación , Hernia Abdominal/cirugía , Piperidinas/administración & dosificación , Propofol/administración & dosificación , Puntos de Acupuntura , Anciano , Anestesia General , Procedimientos Quirúrgicos Electivos , Electroencefalografía , Humanos , Remifentanilo
20.
Nature ; 543(7644): 265-269, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28241141

RESUMEN

Cancer cells are characterized by aberrant epigenetic landscapes and often exploit chromatin machinery to activate oncogenic gene expression programs. Recognition of modified histones by 'reader' proteins constitutes a key mechanism underlying these processes; therefore, targeting such pathways holds clinical promise, as exemplified by the development of bromodomain and extra-terminal (BET) inhibitors. We recently identified the YEATS domain as an acetyl-lysine-binding module, but its functional importance in human cancer remains unknown. Here we show that the YEATS domain-containing protein ENL, but not its paralogue AF9, is required for disease maintenance in acute myeloid leukaemia. CRISPR-Cas9-mediated depletion of ENL led to anti-leukaemic effects, including increased terminal myeloid differentiation and suppression of leukaemia growth in vitro and in vivo. Biochemical and crystal structural studies and chromatin-immunoprecipitation followed by sequencing analyses revealed that ENL binds to acetylated histone H3, and co-localizes with H3K27ac and H3K9ac on the promoters of actively transcribed genes that are essential for leukaemia. Disrupting the interaction between the YEATS domain and histone acetylation via structure-based mutagenesis reduced the recruitment of RNA polymerase II to ENL-target genes, leading to the suppression of oncogenic gene expression programs. Notably, disrupting the functionality of ENL further sensitized leukaemia cells to BET inhibitors. Together, our data identify ENL as a histone acetylation reader that regulates oncogenic transcriptional programs in acute myeloid leukaemia, and suggest that displacement of ENL from chromatin may be a promising epigenetic therapy, alone or in combination with BET inhibitors, for aggressive leukaemia.


Asunto(s)
Acetilación , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Oncogenes/genética , Factores de Elongación Transcripcional/metabolismo , Animales , Sistemas CRISPR-Cas , Línea Celular Tumoral , Epigénesis Genética , Femenino , Edición Génica , Histonas/química , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Lisina/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Dominios Proteicos , ARN Polimerasa II/metabolismo , Transcripción Genética , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/deficiencia , Factores de Elongación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA