Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sci Adv ; 10(23): eadm9589, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38838142

RESUMEN

DNA replication stress (RS) is a widespread phenomenon in carcinogenesis, causing genomic instability and extensive chromatin alterations. DNA damage leads to activation of innate immune signaling, but little is known about transcriptional regulators mediating such signaling upon RS. Using a chemical screen, we identified protein arginine methyltransferase 5 (PRMT5) as a key mediator of RS-dependent induction of interferon-stimulated genes (ISGs). This response is also associated with reactivation of endogenous retroviruses (ERVs). Using quantitative mass spectrometry, we identify proteins with PRMT5-dependent symmetric dimethylarginine (SDMA) modification induced upon RS. Among these, we show that PRMT5 targets and modulates the activity of ZNF326, a zinc finger protein essential for ISG response. Our data demonstrate a role for PRMT5-mediated SDMA in the context of RS-induced transcriptional induction, affecting physiological homeostasis and cancer therapy.


Asunto(s)
Replicación del ADN , Inmunidad Innata , Proteína-Arginina N-Metiltransferasas , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Humanos , Transducción de Señal , Arginina/metabolismo , Arginina/análogos & derivados , Estrés Fisiológico , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Daño del ADN , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
2.
Nat Immunol ; 25(7): 1257-1269, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38806707

RESUMEN

The circadian clock is a critical regulator of immunity, and this circadian control of immune modulation has an essential function in host defense and tumor immunosurveillance. Here we use a single-cell RNA sequencing approach and a genetic model of colorectal cancer to identify clock-dependent changes to the immune landscape that control the abundance of immunosuppressive cells and consequent suppression of cytotoxic CD8+ T cells. Of these immunosuppressive cell types, PD-L1-expressing myeloid-derived suppressor cells (MDSCs) peak in abundance in a rhythmic manner. Disruption of the epithelial cell clock regulates the secretion of cytokines that promote heightened inflammation, recruitment of neutrophils and the subsequent development of MDSCs. We also show that time-of-day anti-PD-L1 delivery is most effective when synchronized with the abundance of immunosuppressive MDSCs. Collectively, these data indicate that circadian gating of tumor immunosuppression informs the timing and efficacy of immune checkpoint inhibitors.


Asunto(s)
Antígeno B7-H1 , Relojes Circadianos , Inhibidores de Puntos de Control Inmunológico , Células Supresoras de Origen Mieloide , Animales , Ratones , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/farmacología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Relojes Circadianos/inmunología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Ratones Endogámicos C57BL , Ritmo Circadiano/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Neoplasias Colorrectales/tratamiento farmacológico , Microambiente Tumoral/inmunología , Tolerancia Inmunológica , Humanos , Femenino , Línea Celular Tumoral , Análisis de la Célula Individual , Terapia de Inmunosupresión , Citocinas/metabolismo , Masculino
3.
Nat Struct Mol Biol ; 30(12): 1947-1957, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38087090

RESUMEN

JTE-607 is an anticancer and anti-inflammatory compound and its active form, compound 2, directly binds to and inhibits CPSF73, the endonuclease for the cleavage step in pre-messenger RNA (pre-mRNA) 3' processing. Surprisingly, compound 2-mediated inhibition of pre-mRNA cleavage is sequence specific and the drug sensitivity is predominantly determined by sequences flanking the cleavage site (CS). Using massively parallel in vitro assays, we identified key sequence features that determine drug sensitivity. We trained a machine learning model that can predict poly(A) site (PAS) relative sensitivity to compound 2 and provide the molecular basis for understanding the impact of JTE-607 on PAS selection and transcription termination genome wide. We propose that CPSF73 and associated factors bind to the CS region in a sequence-dependent manner and the interaction affinity determines compound 2 sensitivity. These results have not only elucidated the mechanism of action of JTE-607, but also unveiled an evolutionarily conserved sequence specificity of the mRNA 3' processing machinery.


Asunto(s)
Precursores del ARN , Procesamiento Postranscripcional del ARN , Línea Celular , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Nat Cancer ; 4(8): 1157-1175, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37537299

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide. ß-Catenin (CTNNB1)-mutated HCC represents 30% of cases of the disease with no precision therapeutics available. Using chemical libraries derived from clinical multi-kinase inhibitor (KI) scaffolds, we screened HCC organoids to identify WNTinib, a KI with exquisite selectivity in CTNNB1-mutated human and murine models, including patient samples. Multiomic and target engagement analyses, combined with rescue experiments and in vitro and in vivo efficacy studies, revealed that WNTinib is superior to clinical KIs and inhibits KIT/mitogen-activated protein kinase (MAPK) signaling at multiple nodes. Moreover, we demonstrate that reduced engagement on BRAF and p38α kinases by WNTinib relative to several multi-KIs is necessary to avoid compensatory feedback signaling-providing a durable and selective transcriptional repression of mutant ß-catenin/Wnt targets through nuclear translocation of the EZH2 transcriptional repressor. Our studies uncover a previously unknown mechanism to harness the KIT/MAPK/EZH2 pathway to potently and selectively antagonize CTNNB1-mutant HCC with an unprecedented wide therapeutic index.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Ratones , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , beta Catenina/genética , beta Catenina/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Factores de Transcripción/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
5.
bioRxiv ; 2023 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-37090613

RESUMEN

JTE-607 is a small molecule compound with anti-inflammation and anti-cancer activities. Upon entering the cell, it is hydrolyzed to Compound 2, which directly binds to and inhibits CPSF73, the endonuclease for the cleavage step in pre-mRNA 3' processing. Although CPSF73 is universally required for mRNA 3' end formation, we have unexpectedly found that Compound 2- mediated inhibition of pre-mRNA 3' processing is sequence-specific and that the sequences flanking the cleavage site (CS) are a major determinant for drug sensitivity. By using massively parallel in vitro assays, we have measured the Compound 2 sensitivities of over 260,000 sequence variants and identified key sequence features that determine drug sensitivity. A machine learning model trained on these data can predict the impact of JTE-607 on poly(A) site (PAS) selection and transcription termination genome-wide. We propose a biochemical model in which CPSF73 and other mRNA 3' processing factors bind to RNA of the CS region in a sequence-specific manner and the affinity of such interaction determines the Compound 2 sensitivity of a PAS. As the Compound 2-resistant CS sequences, characterized by U/A-rich motifs, are prevalent in PASs from yeast to human, the CS region sequence may have more fundamental functions beyond determining drug resistance. Together, our study not only characterized the mechanism of action of a compound with clinical implications, but also revealed a previously unknown and evolutionarily conserved sequence-specificity of the mRNA 3' processing machinery.

6.
Nature ; 606(7916): 945-952, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35732742

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a heterogenous neurodegenerative disorder that affects motor neurons and voluntary muscle control1. ALS heterogeneity includes the age of manifestation, the rate of progression and the anatomical sites of symptom onset. Disease-causing mutations in specific genes have been identified and define different subtypes of ALS1. Although several ALS-associated genes have been shown to affect immune functions2, whether specific immune features account for ALS heterogeneity is poorly understood. Amyotrophic lateral sclerosis-4 (ALS4) is characterized by juvenile onset and slow progression3. Patients with ALS4 show motor difficulties by the time that they are in their thirties, and most of them require devices to assist with walking by their fifties. ALS4 is caused by mutations in the senataxin gene (SETX). Here, using Setx knock-in mice that carry the ALS4-causative L389S mutation, we describe an immunological signature that consists of clonally expanded, terminally differentiated effector memory (TEMRA) CD8 T cells in the central nervous system and the blood of knock-in mice. Increased frequencies of antigen-specific CD8 T cells in knock-in mice mirror the progression of motor neuron disease and correlate with anti-glioma immunity. Furthermore, bone marrow transplantation experiments indicate that the immune system has a key role in ALS4 neurodegeneration. In patients with ALS4, clonally expanded TEMRA CD8 T cells circulate in the peripheral blood. Our results provide evidence of an antigen-specific CD8 T cell response in ALS4, which could be used to unravel disease mechanisms and as a potential biomarker of disease state.


Asunto(s)
Esclerosis Amiotrófica Lateral , Linfocitos T CD8-positivos , Células Clonales , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/patología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Células Clonales/patología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Técnicas de Sustitución del Gen , Ratones , Neuronas Motoras/patología , Enzimas Multifuncionales/genética , Enzimas Multifuncionales/metabolismo , Mutación , ARN Helicasas/genética , ARN Helicasas/metabolismo
7.
Elife ; 102021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34075878

RESUMEN

High spliceosome activity is a dependency for cancer cells, making them more vulnerable to perturbation of the splicing machinery compared to normal cells. To identify splicing factors important for prostate cancer (PCa) fitness, we performed pooled shRNA screens in vitro and in vivo. Our screens identified heterogeneous nuclear ribonucleoprotein M (HNRNPM) as a regulator of PCa cell growth. RNA- and eCLIP-sequencing identified HNRNPM binding to transcripts of key homeostatic genes. HNRNPM binding to its targets prevents aberrant exon inclusion and backsplicing events. In both linear and circular mis-spliced transcripts, HNRNPM preferentially binds to GU-rich elements in long flanking proximal introns. Mimicry of HNRNPM-dependent linear-splicing events using splice-switching-antisense-oligonucleotides was sufficient to inhibit PCa cell growth. This suggests that PCa dependence on HNRNPM is likely a result of mis-splicing of key homeostatic coding and non-coding genes. Our results have further been confirmed in other solid tumors. Taken together, our data reveal a role for HNRNPM in supporting cancer cell fitness. Inhibition of HNRNPM activity is therefore a potential therapeutic strategy in suppressing growth of PCa and other solid tumors.


Asunto(s)
Adenocarcinoma/metabolismo , Proliferación Celular , Ribonucleoproteína Heterogénea-Nuclear Grupo M/metabolismo , Neoplasias de la Próstata/metabolismo , Empalme del ARN , ARN Circular/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Ribonucleoproteína Heterogénea-Nuclear Grupo M/genética , Humanos , Masculino , Ratones SCID , Células PC-3 , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Circular/genética , Carga Tumoral , Células Tumorales Cultivadas
8.
Cell ; 175(7): 1931-1945.e18, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30550790

RESUMEN

Mosquito-borne flaviviruses, including dengue virus (DENV) and Zika virus (ZIKV), are a growing public health concern. Systems-level analysis of how flaviviruses hijack cellular processes through virus-host protein-protein interactions (PPIs) provides information about their replication and pathogenic mechanisms. We used affinity purification-mass spectrometry (AP-MS) to compare flavivirus-host interactions for two viruses (DENV and ZIKV) in two hosts (human and mosquito). Conserved virus-host PPIs revealed that the flavivirus NS5 protein suppresses interferon stimulated genes by inhibiting recruitment of the transcription complex PAF1C and that chemical modulation of SEC61 inhibits DENV and ZIKV replication in human and mosquito cells. Finally, we identified a ZIKV-specific interaction between NS4A and ANKLE2, a gene linked to hereditary microcephaly, and showed that ZIKV NS4A causes microcephaly in Drosophila in an ANKLE2-dependent manner. Thus, comparative flavivirus-host PPI mapping provides biological insights and, when coupled with in vivo models, can be used to unravel pathogenic mechanisms.


Asunto(s)
Virus del Dengue , Dengue , Proteínas de la Membrana , Proteínas Nucleares , Proteínas no Estructurales Virales , Infección por el Virus Zika , Virus Zika , Animales , Línea Celular Tumoral , Culicidae , Dengue/genética , Dengue/metabolismo , Dengue/patología , Virus del Dengue/genética , Virus del Dengue/metabolismo , Virus del Dengue/patogenicidad , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mapeo de Interacción de Proteínas , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Virus Zika/genética , Virus Zika/metabolismo , Virus Zika/patogenicidad , Infección por el Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/patología
9.
Immunity ; 49(5): 819-828.e6, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30413362

RESUMEN

Inducing graft acceptance without chronic immunosuppression remains an elusive goal in organ transplantation. Using an experimental transplantation mouse model, we demonstrate that local macrophage activation through dectin-1 and toll-like receptor 4 (TLR4) drives trained immunity-associated cytokine production during allograft rejection. We conducted nanoimmunotherapeutic studies and found that a short-term mTOR-specific high-density lipoprotein (HDL) nanobiologic treatment (mTORi-HDL) averted macrophage aerobic glycolysis and the epigenetic modifications underlying inflammatory cytokine production. The resulting regulatory macrophages prevented alloreactive CD8+ T cell-mediated immunity and promoted tolerogenic CD4+ regulatory T (Treg) cell expansion. To enhance therapeutic efficacy, we complemented the mTORi-HDL treatment with a CD40-TRAF6-specific nanobiologic (TRAF6i-HDL) that inhibits co-stimulation. This synergistic nanoimmunotherapy resulted in indefinite allograft survival. Together, we show that HDL-based nanoimmunotherapy can be employed to control macrophage function in vivo. Our strategy, focused on preventing inflammatory innate immune responses, provides a framework for developing targeted therapies that promote immunological tolerance.


Asunto(s)
Supervivencia de Injerto/inmunología , Terapia de Inmunosupresión , Inflamación/inmunología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Trasplante de Órganos , Aloinjertos , Animales , Biomarcadores , Proteína HMGB1/genética , Tolerancia Inmunológica , Inmunidad Innata , Memoria Inmunológica , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Vimentina/genética
10.
Cell ; 174(6): 1522-1536.e22, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146161

RESUMEN

How transcription affects genome 3D organization is not well understood. We found that during influenza A (IAV) infection, rampant transcription rapidly reorganizes host cell chromatin interactions. These changes occur at the ends of highly transcribed genes, where global inhibition of transcription termination by IAV NS1 protein causes readthrough transcription for hundreds of kilobases. In these readthrough regions, elongating RNA polymerase II disrupts chromatin interactions by inducing cohesin displacement from CTCF sites, leading to locus decompaction. Readthrough transcription into heterochromatin regions switches them from the inert (B) to the permissive (A) chromatin compartment and enables transcription factor binding. Data from non-viral transcription stimuli show that transcription similarly affects cohesin-mediated chromatin contacts within gene bodies. Conversely, inhibition of transcription elongation allows cohesin to accumulate at previously transcribed intragenic CTCF sites and to mediate chromatin looping and compaction. Our data indicate that transcription elongation by RNA polymerase II remodels genome 3D architecture.


Asunto(s)
Cromatina/metabolismo , Genoma Humano , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Sitios de Unión , Factor de Unión a CCCTC/química , Factor de Unión a CCCTC/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Cromatina/química , Proteínas Cromosómicas no Histona/metabolismo , Flavonoides/farmacología , Humanos , Interferón beta/farmacología , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/virología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Piperidinas/farmacología , Unión Proteica , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Polimerasa II/genética , ARN Polimerasa II/metabolismo , ARN Interferente Pequeño/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Cohesinas
11.
Nat Rev Mol Cell Biol ; 19(4): 245-261, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29184195

RESUMEN

Multiple cell-signalling pathways converge on chromatin to induce gene expression programmes. The inducible transcriptional programmes that are established as a result of inflammatory or oncogenic signals are controlled by shared chromatin regulators. Therapeutic targeting of such chromatin dependencies has proved effective for controlling tumorigenesis and for preventing immunopathologies that are driven by overt inflammation. In this Review, we discuss how chromatin dependencies are established to regulate the expression of key oncogenes and inflammation-promoting genes and how a better mechanistic understanding of such chromatin dependencies can be leveraged to improve the magnitude, timing, duration and selectivity of cell responses with the aim of minimizing unwanted cellular and systemic effects. Recently, exciting progress has been made in cancer immunotherapy and in the development of drugs that target chromatin regulators. We discuss recent advances in clinical trials and the challenge of combining immune-cell-based therapies and epigenetic therapies to improve human health.


Asunto(s)
Cromatina/genética , Inflamación/genética , Neoplasias/genética , Animales , Carcinogénesis/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ensamble y Desensamble de Cromatina/genética , Epigénesis Genética , Expresión Génica/efectos de los fármacos , Terapia Genética , Humanos , Inflamación/metabolismo , Modelos Genéticos , Neoplasias/metabolismo , Neoplasias/terapia , Transducción de Señal/genética , Factores de Transcripción/metabolismo
12.
Immunity ; 44(1): 46-58, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26789921

RESUMEN

Viruses are obligate parasites and thus require the machinery of the host cell to replicate. Inhibition of host factors co-opted during active infection is a strategy hosts use to suppress viral replication and a potential pan-antiviral therapy. To define the cellular proteins and processes required for a virus during infection is thus crucial to understanding the mechanisms of virally induced disease. In this report, we generated fully infectious tagged influenza viruses and used infection-based proteomics to identify pivotal arms of cellular signaling required for influenza virus growth and infectivity. Using mathematical modeling and genetic and pharmacologic approaches, we revealed that modulation of Sec61-mediated cotranslational translocation selectively impaired glycoprotein proteostasis of influenza as well as HIV and dengue viruses and led to inhibition of viral growth and infectivity. Thus, by studying virus-human protein-protein interactions in the context of active replication, we have identified targetable host factors for broad-spectrum antiviral therapies.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Virus de la Influenza A/fisiología , Virus de la Influenza A/patogenicidad , Modelos Teóricos , Replicación Viral/fisiología , Virus del Dengue/patogenicidad , Virus del Dengue/fisiología , VIH/patogenicidad , VIH/fisiología , Humanos , Inmunoprecipitación , Espectrometría de Masas , Pliegue de Proteína , Proteómica
13.
Nat Immunol ; 16(5): 485-94, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25822250

RESUMEN

The human helicase senataxin (SETX) has been linked to the neurodegenerative diseases amyotrophic lateral sclerosis (ALS4) and ataxia with oculomotor apraxia (AOA2). Here we identified a role for SETX in controlling the antiviral response. Cells that had undergone depletion of SETX and SETX-deficient cells derived from patients with AOA2 had higher expression of antiviral mediators in response to infection than did wild-type cells. Mechanistically, we propose a model whereby SETX attenuates the activity of RNA polymerase II (RNAPII) at genes stimulated after a virus is sensed and thus controls the magnitude of the host response to pathogens and the biogenesis of various RNA viruses (e.g., influenza A virus and West Nile virus). Our data indicate a potentially causal link among inborn errors in SETX, susceptibility to infection and the development of neurologic disorders.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Gripe Humana/inmunología , Orthomyxoviridae/fisiología , ARN Helicasas/metabolismo , ARN Polimerasa II/metabolismo , Degeneraciones Espinocerebelosas/genética , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/fisiología , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Citocinas/metabolismo , ADN Helicasas , Perros , Regulación hacia Abajo , Humanos , Inmunidad Innata/genética , Factor 3 Regulador del Interferón/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Análisis por Micromatrices , Enzimas Multifuncionales , ARN Helicasas/genética , ARN Polimerasa II/genética , ARN Interferente Pequeño/genética , Ataxias Espinocerebelosas/congénito , Células Vero , Replicación Viral/genética
14.
Nature ; 468(7327): 1119-23, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21068722

RESUMEN

Interaction of pathogens with cells of the immune system results in activation of inflammatory gene expression. This response, although vital for immune defence, is frequently deleterious to the host due to the exaggerated production of inflammatory proteins. The scope of inflammatory responses reflects the activation state of signalling proteins upstream of inflammatory genes as well as signal-induced assembly of nuclear chromatin complexes that support mRNA expression. Recognition of post-translationally modified histones by nuclear proteins that initiate mRNA transcription and support mRNA elongation is a critical step in the regulation of gene expression. Here we present a novel pharmacological approach that targets inflammatory gene expression by interfering with the recognition of acetylated histones by the bromodomain and extra terminal domain (BET) family of proteins. We describe a synthetic compound (I-BET) that by 'mimicking' acetylated histones disrupts chromatin complexes responsible for the expression of key inflammatory genes in activated macrophages, and confers protection against lipopolysaccharide-induced endotoxic shock and bacteria-induced sepsis. Our findings suggest that synthetic compounds specifically targeting proteins that recognize post-translationally modified histones can serve as a new generation of immunomodulatory drugs.


Asunto(s)
Antiinflamatorios/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Inflamación , Macrófagos/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Antiinflamatorios/química , Antiinflamatorios/uso terapéutico , Benzodiazepinas , Células Cultivadas , Epigenómica , Estudio de Asociación del Genoma Completo , Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Estimación de Kaplan-Meier , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/fisiopatología , Infecciones por Salmonella/prevención & control , Salmonella typhimurium , Sepsis/tratamiento farmacológico , Sepsis/prevención & control , Choque Séptico/tratamiento farmacológico , Choque Séptico/prevención & control
15.
J Exp Med ; 200(1): 107-13, 2004 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15226358

RESUMEN

Transcription factors of the nuclear factor (NF)-kappaB/Rel family translocate into the nucleus upon degradation of the IkappaBs. Postinduction repression of NF-kappaB activity depends on NF-kappaB-regulated resynthesis of IkappaBalpha, which dissociates NF-kappaB from DNA and exports it to the cytosol. We found that after activation, p65/RelA is degraded by the proteasome in the nucleus and in a DNA binding-dependent manner. If proteasome activity is blocked, NF-kappaB is not promptly removed from some target genes in spite of IkappaBalpha resynthesis and sustained transcription occurs. These results indicate that proteasomal degradation of p65/RelA does not merely regulate its stability and abundance, but also actively promotes transcriptional termination.


Asunto(s)
Regulación de la Expresión Génica , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Células 3T3 , Transporte Activo de Núcleo Celular/fisiología , Animales , Cisteína Endopeptidasas/metabolismo , Humanos , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Ratones , Complejos Multienzimáticos/metabolismo , Inhibidor NF-kappaB alfa , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Factor de Transcripción ReIA , Transcripción Genética , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA