Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(11): 113322, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37883227

RESUMEN

Fibrosis, characterized by sustained activation of myofibroblasts and excessive extracellular matrix (ECM) deposition, is known to be associated with chronic inflammation. Receptor-interacting protein kinase 3 (RIPK3), the central kinase of necroptosis signaling, is upregulated in fibrosis and contributes to tumor necrosis factor (TNF)-mediated inflammation. In bile-duct-ligation-induced liver fibrosis, we found that myofibroblasts are the major cell type expressing RIPK3. Genetic ablation of ß1 integrin, the major profibrotic ECM receptor in fibroblasts, not only abolished ECM fibrillogenesis but also blunted RIPK3 expression via a mechanism mediated by the chromatin-remodeling factor chromodomain helicase DNA-binding protein 4 (CHD4). While the function of CHD4 has been conventionally linked to the nucleosome-remodeling deacetylase (NuRD) and CHD4-ADNP-HP1(ChAHP) complexes, we found that CHD4 potently repressed a set of genes, including Ripk3, with high locus specificity but independent of either the NuRD or the ChAHP complex. Thus, our data uncover that ß1 integrin intrinsically links fibrotic signaling to RIPK3-driven inflammation via a novel mode of action of CHD4.


Asunto(s)
Integrina beta1 , Necroptosis , Humanos , Integrina beta1/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Factores de Transcripción/genética , Nucleosomas , Fibrosis , Inflamación
2.
Nat Cancer ; 4(10): 1508-1525, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37723306

RESUMEN

The PDCD1-encoded immune checkpoint receptor PD-1 is a key tumor suppressor in T cells that is recurrently inactivated in T cell non-Hodgkin lymphomas (T-NHLs). The highest frequencies of PDCD1 deletions are detected in advanced disease, predicting inferior prognosis. However, the tumor-suppressive mechanisms of PD-1 signaling remain unknown. Here, using tractable mouse models for T-NHL and primary patient samples, we demonstrate that PD-1 signaling suppresses T cell malignancy by restricting glycolytic energy and acetyl coenzyme A (CoA) production. In addition, PD-1 inactivation enforces ATP citrate lyase (ACLY) activity, which generates extramitochondrial acetyl-CoA for histone acetylation to enable hyperactivity of activating protein 1 (AP-1) transcription factors. Conversely, pharmacological ACLY inhibition impedes aberrant AP-1 signaling in PD-1-deficient T-NHLs and is toxic to these cancers. Our data uncover genotype-specific vulnerabilities in PDCD1-mutated T-NHL and identify PD-1 as regulator of AP-1 activity.


Asunto(s)
Linfoma de Células T Periférico , Linfoma de Células T , Ratones , Animales , Humanos , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Linfoma de Células T/genética , Genes Supresores de Tumor , Acetilcoenzima A/metabolismo , Glucólisis/genética
3.
Cell Rep ; 42(6): 112533, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37257449

RESUMEN

The acquisition of mesenchymal traits is considered a hallmark of breast cancer progression. However, the functional relevance of epithelial-to-mesenchymal transition (EMT) remains controversial and context dependent. Here, we isolate epithelial and mesenchymal populations from human breast cancer metastatic biopsies and assess their functional potential in vivo. Strikingly, progressively decreasing epithelial cell adhesion molecule (EPCAM) levels correlate with declining disease propagation. Mechanistically, we find that persistent EPCAM expression marks epithelial clones that resist EMT induction and propagate competitively. In contrast, loss of EPCAM defines clones arrested in a mesenchymal state, with concomitant suppression of tumorigenicity and metastatic potential. This dichotomy results from distinct clonal trajectories impacting global epigenetic programs that are determined by the interplay between human ZEB1 and its target GRHL2. Collectively, our results indicate that susceptibility to irreversible EMT restrains clonal propagation, whereas resistance to mesenchymal reprogramming sustains disease spread in multiple models of human metastatic breast cancer, including patient-derived cells in vivo.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Molécula de Adhesión Celular Epitelial , Neoplasias de la Mama/patología , Línea Celular Tumoral , Mama/metabolismo , Células Clonales/metabolismo , Transición Epitelial-Mesenquimal
4.
Nucleic Acids Res ; 51(10): 5144-5161, 2023 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-37021550

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most prevalent neuromuscular disorders. The disease is linked to copy number reduction and/or epigenetic alterations of the D4Z4 macrosatellite on chromosome 4q35 and associated with aberrant gain of expression of the transcription factor DUX4, which triggers a pro-apoptotic transcriptional program leading to muscle wasting. As today, no cure or therapeutic option is available to FSHD patients. Given its centrality in FSHD, blocking DUX4 expression with small molecule drugs is an attractive option. We previously showed that the long non protein-coding RNA DBE-T is required for aberrant DUX4 expression in FSHD. Using affinity purification followed by proteomics, here we identified the chromatin remodeling protein WDR5 as a novel DBE-T interactor and a key player required for the biological activity of the lncRNA. We found that WDR5 is required for the expression of DUX4 and its targets in primary FSHD muscle cells. Moreover, targeting WDR5 rescues both cell viability and myogenic differentiation of FSHD patient cells. Notably, comparable results were obtained by pharmacological inhibition of WDR5. Importantly, WDR5 targeting was safe to healthy donor muscle cells. Our results support a pivotal role of WDR5 in the activation of DUX4 expression identifying a druggable target for an innovative therapeutic approach for FSHD.


Asunto(s)
Distrofia Muscular Facioescapulohumeral , Humanos , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Células Musculares/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapulohumeral/metabolismo , Factores de Transcripción/metabolismo
5.
Nat Commun ; 13(1): 5447, 2022 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-36123357

RESUMEN

Silencing of endogenous retroviruses (ERVs) is largely mediated by repressive chromatin modifications H3K9me3 and DNA methylation. On ERVs, these modifications are mainly deposited by the histone methyltransferase Setdb1 and by the maintenance DNA methyltransferase Dnmt1. Knock-out of either Setdb1 or Dnmt1 leads to ERV de-repression in various cell types. However, it is currently not known if H3K9me3 and DNA methylation depend on each other for ERV silencing. Here we show that conditional knock-out of Setdb1 in mouse embryonic endoderm results in ERV de-repression in visceral endoderm (VE) descendants and does not occur in definitive endoderm (DE). Deletion of Setdb1 in VE progenitors results in loss of H3K9me3 and reduced DNA methylation of Intracisternal A-particle (IAP) elements, consistent with up-regulation of this ERV family. In DE, loss of Setdb1 does not affect H3K9me3 nor DNA methylation, suggesting Setdb1-independent pathways for maintaining these modifications. Importantly, Dnmt1 knock-out results in IAP de-repression in both visceral and definitive endoderm cells, while H3K9me3 is unaltered. Thus, our data suggest a dominant role of DNA methylation over H3K9me3 for IAP silencing in endoderm cells. Our findings suggest that Setdb1-meditated H3K9me3 is not sufficient for IAP silencing, but rather critical for maintaining high DNA methylation.


Asunto(s)
Metilación de ADN , Retrovirus Endógenos , Animales , Cromatina/metabolismo , ADN/metabolismo , Endodermo/metabolismo , Retrovirus Endógenos/metabolismo , Histona Metiltransferasas/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones
6.
Nat Commun ; 13(1): 1861, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35387992

RESUMEN

The mammalian epigenome contains thousands of heterochromatin nanodomains (HNDs) marked by di- and trimethylation of histone H3 at lysine 9 (H3K9me2/3), which have a typical size of 3-10 nucleosomes. However, what governs HND location and extension is only partly understood. Here, we address this issue by introducing the chromatin hierarchical lattice framework (ChromHL) that predicts chromatin state patterns with single-nucleotide resolution. ChromHL is applied to analyse four HND types in mouse embryonic stem cells that are defined by histone methylases SUV39H1/2 or GLP, transcription factor ADNP or chromatin remodeller ATRX. We find that HND patterns can be computed from PAX3/9, ADNP and LINE1 sequence motifs as nucleation sites and boundaries that are determined by DNA sequence (e.g. CTCF binding sites), cooperative interactions between nucleosomes as well as nucleosome-HP1 interactions. Thus, ChromHL rationalizes how patterns of H3K9me2/3 are established and changed via the activity of protein factors in processes like cell differentiation.


Asunto(s)
Heterocromatina , Nucleosomas , Animales , Secuencia de Bases , Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/genética , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodominio/genética , Ratones , Proteínas del Tejido Nervioso/metabolismo , Nucleosomas/genética
7.
Exp Hematol ; 108: 26-35, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35181392

RESUMEN

GATA2 zinc-finger (ZF) mutations are associated with distinct entities of myeloid malignancies. The specific distribution of these mutations points toward different mechanisms of leukemogenesis depending on the ZF domain affected. In this study, we compared recurring somatic mutations in ZF1 and ZF2. All tested ZF mutants disrupted DNA binding in vitro. In transcription assays, co-expression of FOG1 counteracted GATA2-dependent transcriptional activation, while a variable response to FOG1-mediated repression was observed for individual GATA2 mutants. In primary murine bone marrow cells, GATA2 wild-type (WT) expression inhibited colony formation, while this effect was reduced for both mutants A318T (ZF1) and L359V (ZF2) with a shift toward granulopoiesis. In primary human CD34+ bone marrow cells and in the myeloid cell line K562, ectopic expression of GATA2 L359V, but not A318T or G320D, caused a block of erythroid differentiation accompanied by downregulation of GATA1, STAT5B, and PLCG1. Our findings may explain the role of GATA2 L359V during the progression of chronic myeloid leukemia and the collaboration of GATA2 ZF1 alterations with CEBPA double mutations in erythroleukemia.


Asunto(s)
Factor de Transcripción GATA2 , Leucemia Eritroblástica Aguda , Leucemia Mieloide , Animales , Diferenciación Celular/genética , Factor de Transcripción GATA2/genética , Humanos , Células K562 , Leucemia Eritroblástica Aguda/genética , Ratones , Mutación , Dedos de Zinc
8.
Cancer Res ; 82(4): 695-707, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34903606

RESUMEN

The mortality of patients with pancreatic ductal adenocarcinoma (PDAC) is strongly associated with metastasis, a multistep process that is incompletely understood in this disease. Although genetic drivers of PDAC metastasis have not been defined, transcriptional and epigenetic rewiring can contribute to the metastatic process. The epigenetic eraser histone deacetylase 2 (HDAC2) has been connected to less differentiated PDAC, but the function of HDAC2 in PDAC has not been comprehensively evaluated. Using genetically defined models, we show that HDAC2 is a cellular fitness factor that controls cell cycle in vitro and metastasis in vivo, particularly in undifferentiated, mesenchymal PDAC cells. Unbiased expression profiling detected a core set of HDAC2-regulated genes. HDAC2 controlled expression of several prosurvival receptor tyrosine kinases connected to mesenchymal PDAC, including PDGFRα, PDGFRß, and EGFR. The HDAC2-maintained program disabled the tumor-suppressive arm of the TGFß pathway, explaining impaired metastasis formation of HDAC2-deficient PDAC. These data identify HDAC2 as a tractable player in the PDAC metastatic cascade. The complexity of the function of epigenetic regulators like HDAC2 implicates that an increased understanding of these proteins is needed for implementation of effective epigenetic therapies. SIGNIFICANCE: HDAC2 has a context-specific role in undifferentiated PDAC and the capacity to disseminate systemically, implicating HDAC2 as targetable protein to prevent metastasis.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 2/genética , Neoplasias Pancreáticas/genética , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Histona Desacetilasa 2/metabolismo , Humanos , Estimación de Kaplan-Meier , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transducción de Señal/genética
9.
Nat Commun ; 12(1): 5996, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650047

RESUMEN

Endogenous retroviruses (ERVs) comprise a significant portion of mammalian genomes. Although specific ERV loci feature regulatory roles for host gene expression, most ERV integrations are transcriptionally repressed by Setdb1-mediated H3K9me3 and DNA methylation. However, the protein network which regulates the deposition of these chromatin modifications is still incompletely understood. Here, we perform a genome-wide single guide RNA (sgRNA) screen for genes involved in ERV silencing and identify the GHKL ATPase protein Morc3 as a top-scoring hit. Morc3 knock-out (ko) cells display de-repression, reduced H3K9me3, and increased chromatin accessibility of distinct ERV families. We find that the Morc3 ATPase cycle and Morc3 SUMOylation are important for ERV chromatin regulation. Proteomic analyses reveal that Morc3 mutant proteins fail to interact with the histone H3.3 chaperone Daxx. This interaction depends on Morc3 SUMOylation and Daxx SUMO binding. Notably, in Morc3 ko cells, we observe strongly reduced histone H3.3 on Morc3 binding sites. Thus, our data demonstrate Morc3 as a critical regulator of Daxx-mediated histone H3.3 incorporation to ERV regions.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas Co-Represoras/genética , Proteínas de Unión al ADN/genética , Retrovirus Endógenos/genética , Silenciador del Gen , Chaperonas Moleculares/genética , Adenosina Trifosfatasas/metabolismo , Animales , Línea Celular , Cromatina , Proteínas Co-Represoras/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Retrovirus Endógenos/metabolismo , Técnicas de Inactivación de Genes , N-Metiltransferasa de Histona-Lisina , Histonas/genética , Histonas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Unión Proteica , Proteómica , Sumoilación
11.
Nat Commun ; 12(1): 3885, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162892

RESUMEN

Cell identity is maintained by activation of cell-specific gene programs, regulated by epigenetic marks, transcription factors and chromatin organization. DNA G-quadruplex (G4)-folded regions in cells were reported to be associated with either increased or decreased transcriptional activity. By G4-ChIP-seq/RNA-seq analysis on liposarcoma cells we confirmed that G4s in promoters are invariably associated with high transcription levels in open chromatin. Comparing G4 presence, location and transcript levels in liposarcoma cells to available data on keratinocytes, we showed that the same promoter sequences of the same genes in the two cell lines had different G4-folding state: high transcript levels consistently associated with G4-folding. Transcription factors AP-1 and SP1, whose binding sites were the most significantly represented in G4-folded sequences, coimmunoprecipitated with their G4-folded promoters. Thus, G4s and their associated transcription factors cooperate to determine cell-specific transcriptional programs, making G4s to strongly emerge as new epigenetic regulators of the transcription machinery.


Asunto(s)
G-Cuádruplex , Perfilación de la Expresión Génica/métodos , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Línea Celular Tumoral , ADN/química , ADN/genética , ADN/metabolismo , Humanos , Conformación de Ácido Nucleico , Unión Proteica , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción AP-1/metabolismo
12.
Nat Cell Biol ; 23(7): 692-703, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34168324

RESUMEN

It is generally accepted that epiblast cells ingress into the primitive streak by epithelial-to-mesenchymal transition (EMT) to give rise to the mesoderm; however, it is less clear how the endoderm acquires an epithelial fate. Here, we used embryonic stem cell and mouse embryo knock-in reporter systems to combine time-resolved lineage labelling with high-resolution single-cell transcriptomics. This allowed us to resolve the morphogenetic programs that segregate the mesoderm from the endoderm germ layer. Strikingly, while the mesoderm is formed by classical EMT, the endoderm is formed independent of the key EMT transcription factor Snail1 by mechanisms of epithelial cell plasticity. Importantly, forkhead box transcription factor A2 (Foxa2) acts as an epithelial gatekeeper and EMT suppressor to shield the endoderm from undergoing a mesenchymal transition. Altogether, these results not only establish the morphogenetic details of germ layer formation, but also have broader implications for stem cell differentiation and cancer metastasis.


Asunto(s)
Blastocisto/fisiología , Plasticidad de la Célula , Endodermo/fisiología , Células Epiteliales/fisiología , Transición Epitelial-Mesenquimal , Gastrulación , Células Madre Embrionarias de Ratones/fisiología , Animales , Blastocisto/citología , Blastocisto/metabolismo , Diferenciación Celular , Línea Celular , Endodermo/citología , Endodermo/metabolismo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Ratones , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo , Fenotipo , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Tiempo
13.
iScience ; 24(6): 102660, 2021 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-34169237

RESUMEN

The spatial organization of chromatin is known to be highly dynamic in response to environmental stress. However, it remains unknown how chromatin dynamics contributes to or modulates disease pathogenesis. Here, we show that upon influenza virus infection, the H4K20me3 methyltransferase Suv4-20h2 binds the viral protein NP, which results in the inactivation of Suv4-20h2 and the dissociation of cohesin from Suv4-20h2. Inactivation of Suv4-20h2 by viral infection or genetic deletion allows the formation of an active chromatin loop at the HoxC8-HoxC6 loci coincident with cohesin loading. HoxC8 and HoxC6 proteins in turn enhance viral replication by inhibiting the Wnt-ß-catenin mediated interferon response. Importantly, loss of Suv4-20h2 augments the pathology of influenza infection in vivo. Thus, Suv4-20h2 acts as a safeguard against influenza virus infection by suppressing cohesin-mediated loop formation.

14.
Nucleic Acids Res ; 49(6): 3217-3241, 2021 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-33675667

RESUMEN

Epstein-Barr virus (EBV), a herpes virus also termed HHV 4 and the first identified human tumor virus, establishes a stable, long-term latent infection in human B cells, its preferred host. Upon induction of EBV's lytic phase, the latently infected cells turn into a virus factory, a process that is governed by EBV. In the lytic, productive phase, all herpes viruses ensure the efficient induction of all lytic viral genes to produce progeny, but certain of these genes also repress the ensuing antiviral responses of the virally infected host cells, regulate their apoptotic death or control the cellular transcriptome. We now find that EBV causes previously unknown massive and global alterations in the chromatin of its host cell upon induction of the viral lytic phase and prior to the onset of viral DNA replication. The viral initiator protein of the lytic cycle, BZLF1, binds to >105 binding sites with different sequence motifs in cellular chromatin in a concentration dependent manner implementing a binary molar switch probably to prevent noise-induced erroneous induction of EBV's lytic phase. Concomitant with DNA binding of BZLF1, silent chromatin opens locally as shown by ATAC-seq experiments, while previously wide-open cellular chromatin becomes inaccessible on a global scale within hours. While viral transcripts increase drastically, the induction of the lytic phase results in a massive reduction of cellular transcripts and a loss of chromatin-chromatin interactions of cellular promoters with their distal regulatory elements as shown in Capture-C experiments. Our data document that EBV's lytic cycle induces discrete early processes that disrupt the architecture of host cellular chromatin and repress the cellular epigenome and transcriptome likely supporting the efficient de novo synthesis of this herpes virus.


Asunto(s)
Cromatina/virología , Regulación de la Expresión Génica , Herpesvirus Humano 4/fisiología , Transactivadores/metabolismo , Transcriptoma , Sitios de Unión , Línea Celular , Cromatina/química , Cromatina/metabolismo , ADN/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Humanos
15.
Sci Rep ; 11(1): 5838, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712646

RESUMEN

Chemotherapy resistance is the main impediment in the treatment of acute myeloid leukaemia (AML). Despite rapid advances, the various mechanisms inducing resistance development remain to be defined in detail. Here we report that loss-of-function mutations (LOF) in the histone methyltransferase EZH2 have the potential to confer resistance against the chemotherapeutic agent cytarabine. We identify seven distinct EZH2 mutations leading to loss of H3K27 trimethylation via multiple mechanisms. Analysis of matched diagnosis and relapse samples reveal a heterogenous regulation of EZH2 and a loss of EZH2 in 50% of patients. We confirm that loss of EZH2 induces resistance against cytarabine in the cell lines HEK293T and K562 as well as in a patient-derived xenograft model. Proteomics and transcriptomics analysis reveal that resistance is conferred by upregulation of multiple direct and indirect EZH2 target genes that are involved in apoptosis evasion, augmentation of proliferation and alteration of transmembrane transporter function. Our data indicate that loss of EZH2 results in upregulation of its target genes, providing the cell with a selective growth advantage, which mediates chemotherapy resistance.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación con Pérdida de Función/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Recurrencia Local de Neoplasia/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Nucleic Acids Res ; 49(2): 847-863, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33410915

RESUMEN

Well-differentiated liposarcoma (WDLPS) is a malignant neoplasia hard to diagnose and treat. Its main molecular signature is amplification of the MDM2-containing genomic region. The MDM2 oncogene is the master regulator of p53: its overexpression enhances p53 degradation and inhibits apoptosis, leading to the tumoral phenotype. Here, we show that the MDM2 inducible promoter G-rich region folds into stable G-quadruplexes both in vitro and in vivo and it is specifically recognized by cellular helicases. Cell treatment with G-quadruplex-ligands reduces MDM2 expression and p53 degradation, thus stimulating cancer cell cycle arrest and apoptosis. Structural characterization of the MDM2 G-quadruplex revealed an extraordinarily stable, unique four-tetrad antiparallel dynamic conformation, amenable to selective targeting. These data indicate the feasibility of an out-of-the-box G-quadruplex-targeting approach to defeat WDLPS and all tumours where restoration of wild-type p53 is sought. They also point to G-quadruplex-dependent genomic instability as possible cause of MDM2 expansion and WDLPS tumorigenesis.


Asunto(s)
G-Cuádruplex , Regulación Neoplásica de la Expresión Génica/genética , Liposarcoma/terapia , Terapia Molecular Dirigida , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Neoplasias de los Tejidos Blandos/terapia , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Simulación por Computador , Humanos , Ligandos , Modelos Genéticos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Mapeo de Interacción de Proteínas , Proteolisis , Proteínas Proto-Oncogénicas c-mdm2/biosíntesis , Proteína p53 Supresora de Tumor/metabolismo
17.
Nat Commun ; 12(1): 464, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33469015

RESUMEN

Conventional dendritic cells (cDC) are key activators of naive T cells, and can be targeted in adults to induce adaptive immunity, but in early life are considered under-developed or functionally immature. Here we show that, in early life, when the immune system develops, cDC2 exhibit a dual hematopoietic origin and, like other myeloid and lymphoid cells, develop in waves. Developmentally distinct cDC2 in early life, despite being distinguishable by fate mapping, are transcriptionally and functionally similar. cDC2 in early and adult life, however, are exposed to distinct cytokine environments that shape their transcriptional profile and alter their ability to sense pathogens, secrete cytokines and polarize T cells. We further show that cDC2 in early life, despite being distinct from cDC2 in adult life, are functionally competent and can induce T cell responses. Our results thus highlight the potential of harnessing cDC2 for boosting immunity in early life.


Asunto(s)
Inmunidad Adaptativa/fisiología , Diferenciación Celular/genética , Citocinas/metabolismo , Células Dendríticas/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Factores de Edad , Animales , Diferenciación Celular/inmunología , Separación Celular , Células Dendríticas/metabolismo , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/fisiología , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Cultivo Primario de Células , RNA-Seq , Análisis de la Célula Individual , Linfocitos T/inmunología , Transcriptoma/inmunología
18.
Gut ; 70(3): 485-498, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32503845

RESUMEN

OBJECTIVE: The intestinal epithelium is a rapidly renewing tissue which plays central roles in nutrient uptake, barrier function and the prevention of intestinal inflammation. Control of epithelial differentiation is essential to these processes and is dependent on cell type-specific activity of transcription factors which bind to accessible chromatin. Here, we studied the role of SET Domain Bifurcated Histone Lysine Methyltransferase 1, also known as ESET (SETDB1), a histone H3K9 methyltransferase, in intestinal epithelial homeostasis and IBD. DESIGN: We investigated mice with constitutive and inducible intestinal epithelial deletion of Setdb1, studied the expression of SETDB1 in patients with IBD and mouse models of IBD, and investigated the abundance of SETDB1 variants in healthy individuals and patients with IBD. RESULTS: Deletion of intestinal epithelial Setdb1 in mice was associated with defects in intestinal epithelial differentiation, barrier disruption, inflammation and mortality. Mechanistic studies showed that loss of SETDB1 leads to de-silencing of endogenous retroviruses, DNA damage and intestinal epithelial cell death. Predicted loss-of-function variants in human SETDB1 were considerably less frequently observed than expected, consistent with a critical role of SETDB1 in human biology. While the vast majority of patients with IBD showed unimpaired mucosal SETDB1 expression, comparison of IBD and non-IBD exomes revealed over-representation of individual rare missense variants in SETDB1 in IBD, some of which are predicted to be associated with loss of function and may contribute to the pathogenesis of intestinal inflammation. CONCLUSION: SETDB1 plays an essential role in intestinal epithelial homeostasis. Future work is required to investigate whether rare variants in SETDB1 contribute to the pathogenesis of IBD.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/genética , Enfermedades Inflamatorias del Intestino/genética , Mucosa Intestinal/metabolismo , Animales , Diferenciación Celular , Células Epiteliales/metabolismo , Femenino , Silenciador del Gen , Homeostasis/genética , Humanos , Mutación con Pérdida de Función , Masculino , Ratones
19.
Nucleic Acids Res ; 48(19): 10890-10908, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33021676

RESUMEN

Although endogenous retroviruses (ERVs) are known to harbor cis-regulatory elements, their role in modulating cellular immune responses remains poorly understood. Using an RNA-seq approach, we show that several members of the ERV9 lineage, particularly LTR12C elements, are activated upon HIV-1 infection of primary CD4+ T cells. Intriguingly, HIV-1-induced ERVs harboring transcription start sites are primarily found in the vicinity of immunity genes. For example, HIV-1 infection activates LTR12C elements upstream of the interferon-inducible genes GBP2 and GBP5 that encode for broad-spectrum antiviral factors. Reporter assays demonstrated that these LTR12C elements drive gene expression in primary CD4+ T cells. In line with this, HIV-1 infection triggered the expression of a unique GBP2 transcript variant by activating a cryptic transcription start site within LTR12C. Furthermore, stimulation with HIV-1-induced cytokines increased GBP2 and GBP5 expression in human cells, but not in macaque cells that naturally lack the GBP5 gene and the LTR12C element upstream of GBP2. Finally, our findings suggest that GBP2 and GBP5 have already been active against ancient viral pathogens as they suppress the maturation of the extinct retrovirus HERV-K (HML-2). In summary, our findings uncover how human cells can exploit remnants of once-infectious retroviruses to regulate antiviral gene expression.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Retrovirus Endógenos/genética , Regulación de la Expresión Génica/inmunología , Infecciones por VIH/genética , Regiones Promotoras Genéticas , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/inmunología , Células HEK293 , Infecciones por VIH/inmunología , VIH-1 , Humanos , Macaca mulatta , Subgrupos de Linfocitos T/citología
20.
J Am Soc Nephrol ; 31(2): 257-278, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31932472

RESUMEN

BACKGROUND: Mononuclear phagocytes (MPs), including macrophages, monocytes, and dendritic cells (DCs), are phagocytic cells with important roles in immunity. The developmental origin of kidney DCs has been highly debated because of the large phenotypic overlap between macrophages and DCs in this tissue. METHODS: We used fate mapping, RNA sequencing, flow cytometry, confocal microscopy, and histo-cytometry to assess the origin and phenotypic and functional properties of renal DCs in healthy kidney and of DCs after cisplatin and ischemia reperfusion-induced kidney injury. RESULTS: Adult kidney contains at least four subsets of MPs with prominent Clec9a-expression history indicating a DC origin. We demonstrate that these populations are phenotypically, functionally, and transcriptionally distinct from each other. We also show these kidney MPs exhibit unique age-dependent developmental heterogeneity. Kidneys from newborn mice contain a prominent population of embryonic-derived MHCIInegF4/80hiCD11blow macrophages that express T cell Ig and mucin domain containing 4 (TIM-4) and MER receptor tyrosine kinase (MERTK). These macrophages are replaced within a few weeks after birth by phenotypically similar cells that express MHCII but lack TIM-4 and MERTK. MHCII+F4/80hi cells exhibit prominent Clec9a-expression history in adulthood but not early life, indicating additional age-dependent developmental heterogeneity. In AKI, MHCIInegF4/80hi cells reappear in adult kidneys as a result of MHCII downregulation by resident MHCII+F4/80hi cells, possibly in response to prostaglandin E2 (PGE2). RNA sequencing further suggests MHCII+F4/80hi cells help coordinate the recruitment of inflammatory cells during renal injury. CONCLUSIONS: Distinct developmental programs contribute to renal DC and macrophage populations throughout life, which could have important implications for therapies targeting these cells.


Asunto(s)
Células Dendríticas/inmunología , Riñón/inmunología , Macrófagos/inmunología , Nefritis/inmunología , Lesión Renal Aguda/inmunología , Factores de Edad , Animales , Antígeno CD11b/análisis , Receptor 1 de Quimiocinas CX3C/análisis , Proteínas de Unión al Calcio/análisis , Cisplatino/farmacología , Antígenos de Histocompatibilidad Clase II/análisis , Riñón/efectos de los fármacos , Riñón/metabolismo , Lectinas Tipo C/análisis , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/análisis , Receptores Inmunológicos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...