Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell Stem Cell ; 30(3): 250-263.e6, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36803553

RESUMO

Adenosine deaminase acting on RNA1 (ADAR1) preserves genomic integrity by preventing retroviral integration and retrotransposition during stress responses. However, inflammatory-microenvironment-induced ADAR1p110 to p150 splice isoform switching drives cancer stem cell (CSC) generation and therapeutic resistance in 20 malignancies. Previously, predicting and preventing ADAR1p150-mediated malignant RNA editing represented a significant challenge. Thus, we developed lentiviral ADAR1 and splicing reporters for non-invasive detection of splicing-mediated ADAR1 adenosine-to-inosine (A-to-I) RNA editing activation; a quantitative ADAR1p150 intracellular flow cytometric assay; a selective small-molecule inhibitor of splicing-mediated ADAR1 activation, Rebecsinib, which inhibits leukemia stem cell (LSC) self-renewal and prolongs humanized LSC mouse model survival at doses that spare normal hematopoietic stem and progenitor cells (HSPCs); and pre-IND studies showing favorable Rebecsinib toxicokinetic and pharmacodynamic (TK/PD) properties. Together, these results lay the foundation for developing Rebecsinib as a clinical ADAR1p150 antagonist aimed at obviating malignant microenvironment-driven LSC generation.


Assuntos
Adenosina Desaminase , Células-Tronco Hematopoéticas , Camundongos , Animais , Isoformas de Proteínas , Adenosina Desaminase/genética
2.
Emerg Infect Dis ; 28(9): 1842-1846, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35997543

RESUMO

We conducted a retrospective cohort study that tested 2,000 US military personnel for Coccidioides antibodies in a disease-endemic region. The overall incidence of seroconversion was 0.5 cases/100 person-years; 12.5% of persons who seroconverted had illnesses requiring medical care. No significant association was found between demographic characteristics and seroconversion or disease.


Assuntos
Coccidioidomicose , Militares , California , Coccidioides , Coccidioidomicose/epidemiologia , Coccidioidomicose/etiologia , Humanos , Incidência , Estudos Retrospectivos
3.
Int J Mol Sci ; 23(6)2022 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-35328625

RESUMO

Advanced prostate cancer (PCa) patients with bone metastases are treated with androgen pathway directed therapy (APDT). However, this treatment invariably fails and the cancer becomes castration resistant. To elucidate resistance mechanisms and to provide a more predictive pre-clinical research platform reflecting tumor heterogeneity, we established organoids from a patient-derived xenograft (PDX) model of bone metastatic prostate cancer, PCSD1. APDT-resistant PDX-derived organoids (PDOs) emerged when cultured without androgen or with the anti-androgen, enzalutamide. Transcriptomics revealed up-regulation of neurogenic and steroidogenic genes and down-regulation of DNA repair, cell cycle, circadian pathways and the severe acute respiratory syndrome (SARS)-CoV-2 host viral entry factors, ACE2 and TMPRSS2. Time course analysis of the cell cycle in live cells revealed that enzalutamide induced a gradual transition into a reversible dormant state as shown here for the first time at the single cell level in the context of multi-cellular, 3D living organoids using the Fucci2BL fluorescent live cell cycle tracker system. We show here a new mechanism of castration resistance in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids. These PDX organoids can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.


Assuntos
Neoplasias Ósseas/genética , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/genética , Organoides/metabolismo , Neoplasias de Próstata Resistentes à Castração/genética , Androgênios/farmacologia , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Benzamidas/farmacologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , COVID-19/genética , COVID-19/metabolismo , COVID-19/virologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Nitrilas/farmacologia , Feniltioidantoína/farmacologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Virais/genética , Receptores Virais/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Transplante Heterólogo , Internalização do Vírus
4.
Phys Biol ; 18(6)2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34433159

RESUMO

In this work we use a discrete Markov chain approach combined with network centrality measures to identify and predict the location of active sites in globular proteins. To accomplish this, we use a three-dimensional network of proteinCαatoms as nodes connected through weighted edges which represent the varying interaction degree between protein's atoms. We compute the mean first passage time matrixH= {Hji} for this Markov chain and evaluate the averaged number of steps ⟨Hj⟩ to reach single nodenjin order to identify such residues that, on the average, are at the least distant from every other node. We also carry out a graph theory analysis to evaluate closeness centralityCc, betweenness centralityCband eigenvector centralityCemeasures which provide relevant information about the connectivity structure and topology of theCαprotein networks. Finally we also performed an analysis of equivalent random and regular networks of the same sizeNin terms of the average path lengthLand the average clustering coefficient⟨C⟩comparing these with the corresponding values forCαprotein networks. Our results show that the mean-first passage time matrixHand its related quantity ⟨Hj⟩ together withCc,CbandCecan not only predict with relative high accuracy the location of active sites in globular proteins but also exhibit a high feasibility to use them to predict the existence of new regions in protein's structure to identify new potential binding or catalytic activity or, in some cases, the presence of new allosteric pathways.


Assuntos
Dobramento de Proteína , Proteínas/química , Sítios de Ligação , Análise por Conglomerados , Cadeias de Markov , Ligação Proteica , Mapas de Interação de Proteínas
5.
Cell Rep ; 34(4): 108670, 2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33503434

RESUMO

Inflammation-dependent base deaminases promote therapeutic resistance in many malignancies. However, their roles in human pre-leukemia stem cell (pre-LSC) evolution to acute myeloid leukemia stem cells (LSCs) had not been elucidated. Comparative whole-genome and whole-transcriptome sequencing analyses of FACS-purified pre-LSCs from myeloproliferative neoplasm (MPN) patients reveal APOBEC3C upregulation, an increased C-to-T mutational burden, and hematopoietic stem and progenitor cell (HSPC) proliferation during progression, which can be recapitulated by lentiviral APOBEC3C overexpression. In pre-LSCs, inflammatory splice isoform overexpression coincides with APOBEC3C upregulation and ADAR1p150-induced A-to-I RNA hyper-editing. Pre-LSC evolution to LSCs is marked by STAT3 editing, STAT3ß isoform switching, elevated phospho-STAT3, and increased ADAR1p150 expression, which can be prevented by JAK2/STAT3 inhibition with ruxolitinib or fedratinib or lentiviral ADAR1 shRNA knockdown. Conversely, lentiviral ADAR1p150 expression enhances pre-LSC replating and STAT3 splice isoform switching. Thus, pre-LSC evolution to LSCs is fueled by primate-specific APOBEC3C-induced pre-LSC proliferation and ADAR1-mediated splicing deregulation.


Assuntos
Inflamação/imunologia , Leucemia Mieloide Aguda/fisiopatologia , Proliferação de Células , Humanos , Células-Tronco Neoplásicas/metabolismo
6.
Blood Cancer Discov ; 1(1): 48-67, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32974613

RESUMO

Persistence of drug-resistant quiescent leukemic stem cells (LSC) and impaired natural killer (NK) cell immune response account for relapse of chronic myelogenous leukemia (CML). Inactivation of protein phosphatase 2A (PP2A) is essential for CML-quiescent LSC survival and NK cell antitumor activity. Here we show that MIR300 has antiproliferative and PP2A-activating functions that are dose dependently differentially induced by CCND2/CDK6 and SET inhibition, respectively. MIR300 is upregulated in CML LSCs and NK cells by bone marrow microenvironment (BMM) signals to induce quiescence and impair immune response, respectively. Conversely, BCR-ABL1 downregulates MIR300 in CML progenitors to prevent growth arrest and PP2A-mediated apoptosis. Quiescent LSCs escape apoptosis by upregulating TUG1 long noncoding RNA that uncouples and limits MIR300 function to cytostasis. Genetic and pharmacologic MIR300 modulation and/or PP2A-activating drug treatment restore NK cell activity, inhibit BMM-induced growth arrest, and selectively trigger LSC apoptosis in vitro and in patient-derived xenografts; hence, the importance of MIR300 and PP2A activity for CML development and therapy.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva , MicroRNAs , Humanos , Células Matadoras Naturais , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , MicroRNAs/genética , Células-Tronco Neoplásicas , Inibidores de Proteínas Quinases/metabolismo , Proteína Fosfatase 2/genética , Microambiente Tumoral/genética
7.
Cancer Cell ; 35(1): 81-94.e7, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30612940

RESUMO

Adenosine deaminase associated with RNA1 (ADAR1) deregulation contributes to therapeutic resistance in many malignancies. Here we show that ADAR1-induced hyper-editing in normal human hematopoietic progenitors impairs miR-26a maturation, which represses CDKN1A expression indirectly via EZH2, thereby accelerating cell-cycle transit. However, in blast crisis chronic myeloid leukemia progenitors, loss of EZH2 expression and increased CDKN1A oppose cell-cycle transit. Moreover, A-to-I editing of both the MDM2 regulatory microRNA and its binding site within the 3' UTR region stabilizes MDM2 transcripts, thereby enhancing blast crisis progenitor propagation. These data reveal a dual mechanism governing malignant transformation of progenitors that is predicated on hyper-editing of cell-cycle-regulatory miRNAs and the 3' UTR binding site of tumor suppressor miRNAs.


Assuntos
Adenosina Desaminase/genética , Crise Blástica/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas de Ligação a RNA/genética , Regiões 3' não Traduzidas , Animais , Ciclo Celular , Feminino , Edição de Genes , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Células HEK293 , Humanos , Células K562 , Masculino , Camundongos , Transplante de Neoplasias
8.
Nat Commun ; 8(1): 1922, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29203771

RESUMO

Despite novel therapies, relapse of multiple myeloma (MM) is virtually inevitable. Amplification of chromosome 1q, which harbors the inflammation-responsive RNA editase adenosine deaminase acting on RNA (ADAR)1 gene, occurs in 30-50% of MM patients and portends a poor prognosis. Since adenosine-to-inosine RNA editing has recently emerged as a driver of cancer progression, genomic amplification combined with inflammatory cytokine activation of ADAR1 could stimulate MM progression and therapeutic resistance. Here, we report that high ADAR1 RNA expression correlates with reduced patient survival rates in the MMRF CoMMpass data set. Expression of wild-type, but not mutant, ADAR1 enhances Alu-dependent editing and transcriptional activity of GLI1, a Hedgehog (Hh) pathway transcriptional activator and self-renewal agonist, and promotes immunomodulatory drug resistance in vitro. Finally, ADAR1 knockdown reduces regeneration of high-risk MM in serially transplantable patient-derived xenografts. These data demonstrate that ADAR1 promotes malignant regeneration of MM and if selectively inhibited may obviate progression and relapse.


Assuntos
Adenosina Desaminase/genética , Mieloma Múltiplo/genética , Recidiva Local de Neoplasia/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteína GLI1 em Dedos de Zinco/metabolismo , Adenosina Desaminase/metabolismo , Adulto , Idoso , Animais , Estudos de Casos e Controles , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Técnicas de Silenciamento de Genes , Humanos , Técnicas In Vitro , Masculino , Camundongos , Pessoa de Meia-Idade , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Recidiva Local de Neoplasia/metabolismo , Transplante de Neoplasias , Prognóstico , Edição de RNA/genética , Proteínas de Ligação a RNA/metabolismo
9.
Cell Stem Cell ; 19(2): 177-191, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27292188

RESUMO

Post-transcriptional adenosine-to-inosine RNA editing mediated by adenosine deaminase acting on RNA1 (ADAR1) promotes cancer progression and therapeutic resistance. However, ADAR1 editase-dependent mechanisms governing leukemia stem cell (LSC) generation have not been elucidated. In blast crisis chronic myeloid leukemia (BC CML), we show that increased JAK2 signaling and BCR-ABL1 amplification activate ADAR1. In a humanized BC CML mouse model, combined JAK2 and BCR-ABL1 inhibition prevents LSC self-renewal commensurate with ADAR1 downregulation. Lentiviral ADAR1 wild-type, but not an editing-defective ADAR1(E912A) mutant, induces self-renewal gene expression and impairs biogenesis of stem cell regulatory let-7 microRNAs. Combined RNA sequencing, qRT-PCR, CLIP-ADAR1, and pri-let-7 mutagenesis data suggest that ADAR1 promotes LSC generation via let-7 pri-microRNA editing and LIN28B upregulation. A small-molecule tool compound antagonizes ADAR1's effect on LSC self-renewal in stromal co-cultures and restores let-7 biogenesis. Thus, ADAR1 activation represents a unique therapeutic vulnerability in LSCs with active JAK2 signaling.


Assuntos
Adenosina Desaminase/metabolismo , Autorrenovação Celular , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adenosina Desaminase/genética , Animais , Sequência de Bases , Autorrenovação Celular/genética , Proteínas de Fusão bcr-abl/metabolismo , Regulação Leucêmica da Expressão Gênica , Janus Quinase 2/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Edição de RNA/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais/genética
10.
Sci Rep ; 6: 23885, 2016 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-27041210

RESUMO

While implicated in therapeutic resistance, malignant progenitor cell cycle kinetics have been difficult to quantify in real-time. We developed an efficient lentiviral bicistronic fluorescent, ubiquitination-based cell cycle indicator reporter (Fucci2BL) to image live single progenitors on a defined niche coupled with cell cycle gene expression analysis. We have identified key differences in cell cycle regulatory gene expression and transit times between normal and chronic myeloid leukemia progenitors that may inform cancer stem cell eradication strategies.


Assuntos
Perfilação da Expressão Gênica/métodos , Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico por imagem , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Células-Tronco Neoplásicas/metabolismo , Análise de Sequência de RNA/métodos , Nicho de Células-Tronco , Ciclo Celular , Linhagem Celular , Técnicas de Cocultura , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Humanos , Cinética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Microscopia Confocal , Imagem com Lapso de Tempo
11.
J Chem Theory Comput ; 11(2): 683-93, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26579602

RESUMO

A new strategy to develop force fields for molecular fluids is presented. The intermolecular parameters are fitted to reproduce experimental values of target properties at ambient conditions and also the critical temperature. The partial charges are chosen to match the dielectric constant. The Lennard-Jones parameters, εii and σii, are fitted to reproduce the surface tension at the vapor-liquid interface and the liquid density, respectively. The choice of those properties allows obtaining systematically the final parameters using a small number of simulations. It is shown that the use of surface tension as a target property is better than the choice of heat of vaporization. The method is applied to molecules, from all atoms to a coarse-grained level, such as pyridine, dichloromethane, methanol, and 1-ethyl-3-methylimidazolium tetrafluoroborate (EMIM-BF4) at different temperatures and pressures. The heat of vaporization, radial distribution functions, and self-diffusion coeficient are also calculated.

12.
BMC Res Notes ; 8: 616, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26515650

RESUMO

BACKGROUND: Eukaryotic RNA polymerase II contains a C-terminal repeated domain (CTD) consisting of 52 consensus heptad repeats of Y1S2P3T4S5P6S7 that mediate interactions with many cellular proteins to regulate transcription elongation, RNA processing and chromatin structure. A number of CTD-binding proteins have been identified and the crystal structures of several protein-CTD complexes have demonstrated considerable conformational flexibility of the heptad repeats in those interactions. Furthermore, phosphorylation of the CTD at tyrosine, serine and threonine residues can regulate the CTD-protein interactions. Although the interactions of CTD with specific proteins have been elucidated at the atomic level, the capacity and specificity of the CTD-interactome in mammalian cells is not yet determined. RESULTS: A proteomic study was conducted to examine the mammalian CTD-interactome. We utilized six synthetic peptides each consisting of four consensus CTD-repeats with different combinations of serine and tyrosine phosphorylation as affinity-probes to pull-down nuclear proteins from HeLa cells. The pull-down fractions were then analyzed by MUDPIT mass spectrometry, which identified 100 proteins with the majority from the phospho-CTD pull-downs. Proteins pulled-down by serine-phosphorylated CTD-peptides included those containing the previously defined CTD-interacting domain (CID). Using SILAC mass spectrometry, we showed that the in vivo interaction of RNA polymerase II with the mammalian CID-containing RPRD1B is disrupted by CID mutation. We also showed that the CID from four mammalian proteins interacted with pS2-phosphorylated but not pY1pS2-doubly phosphorylated CTD-peptides. However, we also found proteins that were preferentially pulled-down by pY1pS2- or pY1pS5-doubly phosphorylated CTD-peptides. We prepared an antibody against tyrosine phosphorylated CTD and showed that ionizing radiation (IR) induced a transient increase in CTD tyrosine phosphorylation by immunoblotting. Combining SILAC and IMAC purification of phospho-peptides, we found that IR regulated the phosphorylation at four CTD tyrosine sites in different ways. CONCLUSION: Upon phosphorylation, the 52 repeats of the CTD have the capacity to generate a large number of binding sites for cellular proteins. This study confirms previous findings that serine phosphorylation stimulates whereas tyrosine phosphorylation inhibits the protein-binding activity of the CTD. However, tyrosine phosphorylation of the CTD can also stimulate other CTD-protein interactions. The CTD-peptide affinity pull-down method described here can be adopted to survey the mammalian CTD-interactome in various cell types and under different biological conditions.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Neoplasias/química , Peptídeos/metabolismo , RNA Polimerase II/química , Serina/química , Tirosina/química , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Raios gama , Regulação da Expressão Gênica , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Peptídeos/síntese química , Fosforilação/efeitos da radiação , Ligação Proteica/efeitos da radiação , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteômica , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Alinhamento de Sequência , Serina/metabolismo , Transdução de Sinais , Tirosina/metabolismo
13.
J Transl Med ; 13: 98, 2015 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-25889765

RESUMO

BACKGROUND: Dormant leukemia stem cells (LSC) promote therapeutic resistance and leukemic progression as a result of unbridled activation of stem cell gene expression programs. Thus, we hypothesized that 1) deregulation of the hedgehog (Hh) stem cell self-renewal and cell cycle regulatory pathway would promote dormant human LSC generation and 2) that PF-04449913, a clinical antagonist of the GLI2 transcriptional activator, smoothened (SMO), would enhance dormant human LSC eradication. METHODS: To test these postulates, whole transcriptome RNA sequencing (RNA-seq), microarray, qRT-PCR, stromal co-culture, confocal fluorescence microscopic, nanoproteomic, serial transplantation and cell cycle analyses were performed on FACS purified normal, chronic phase (CP) chronic myeloid leukemia (CML), blast crisis (BC) phase CML progenitors with or without PF-04449913 treatment. RESULTS: Notably, RNA-seq analyses revealed that Hh pathway and cell cycle regulatory gene overexpression correlated with leukemic progression. While lentivirally enforced GLI2 expression enhanced leukemic progenitor dormancy in stromal co-cultures, this was not observed with a mutant GLI2 lacking a transactivation domain, suggesting that GLI2 expression prevented cell cycle transit. Selective SMO inhibition with PF-04449913 in humanized stromal co-cultures and LSC xenografts reduced downstream GLI2 protein and cell cycle regulatory gene expression. Moreover, SMO inhibition enhanced cell cycle transit and sensitized BC LSC to tyrosine kinase inhibition in vivo at doses that spare normal HSC. CONCLUSION: In summary, while GLI2, forms part of a core HH pathway transcriptional regulatory network that promotes human myeloid leukemic progression and dormant LSC generation, selective inhibition with PF-04449913 reduces the dormant LSC burden thereby providing a strong rationale for clinical trials predicated on SMO inhibition in combination with TKIs or chemotherapeutic agents with the ultimate aim of obviating leukemic therapeutic resistance, persistence and progression.


Assuntos
Fatores de Transcrição Kruppel-Like/antagonistas & inibidores , Leucemia/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/antagonistas & inibidores , Animais , Sequência de Bases , Técnicas de Cocultura , Primers do DNA , Sangue Fetal/citologia , Proteínas Hedgehog/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcriptoma , Proteína Gli2 com Dedos de Zinco
14.
Nature ; 461(7260): 114-9, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19675569

RESUMO

TRAF6 is a ubiquitin ligase that is essential for the activation of NF-kappaB and MAP kinases in several signalling pathways, including those emanating from the interleukin 1 and Toll-like receptors. TRAF6 functions together with a ubiquitin-conjugating enzyme complex consisting of UBC13 (also known as UBE2N) and UEV1A (UBE2V1) to catalyse Lys 63-linked polyubiquitination, which activates the TAK1 (also known as MAP3K7) kinase complex. TAK1 in turn phosphorylates and activates IkappaB kinase (IKK), leading to the activation of NF-kappaB. Although several proteins are known to be polyubiquitinated in the IL1R and Toll-like receptor pathways, it is not clear whether ubiquitination of any of these proteins is important for TAK1 or IKK activation. By reconstituting TAK1 activation in vitro using purified proteins, here we show that free Lys 63 polyubiquitin chains, which are not conjugated to any target protein, directly activate TAK1 by binding to the ubiquitin receptor TAB2 (also known as MAP3K7IP2). This binding leads to autophosphorylation and activation of TAK1. Furthermore, we found that unanchored polyubiquitin chains synthesized by TRAF6 and UBCH5C (also known as UBE2D3) activate the IKK complex. Disassembly of the polyubiquitin chains by deubiquitination enzymes prevented TAK1 and IKK activation. These results indicate that unanchored polyubiquitin chains directly activate TAK1 and IKK, suggesting a new mechanism of protein kinase regulation.


Assuntos
Quinase I-kappa B/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Poliubiquitina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Enzima Desubiquitinante CYLD , Ativação Enzimática/efeitos dos fármacos , Células HeLa , Humanos , Interleucina-1beta/farmacologia , Lisina/metabolismo , Fosforilação , Poliubiquitina/biossíntese , Receptores Imunológicos , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Enzimas de Conjugação de Ubiquitina , Ubiquitinação
16.
J Phys Chem B ; 111(8): 2033-44, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17269816

RESUMO

Electrolytes confined by spherical, cylindrical, and slit-like charged nanopores are studied. Results for ionic distribution profiles, pressures of the confined fluid, and absorption isotherms are obtained through the hypernetted chain/mean spherical approximation (HNC/MSA) integral equations theory. In spherical and cylindrical geometries, an inward, non-monotonic behavior of the pressure is found as confinement increases, implying a negative compressibility. The pressure vs volume isotherms resemble liquid-vapor van der Waals-like phase transition diagrams. This effect is correlated with a charge separation inside a spherical pore previously reported (Phys. Rev. Lett., 79, 3656, 1997). Here, the mechanism of charge separation and negative compressibility are explored in detail. When compared with the slit-like pore pressure, important qualitative differences are found.

17.
Mol Cell ; 22(2): 245-57, 2006 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-16603398

RESUMO

The receptor interacting protein kinase 1 (RIP1) is essential for the activation of nuclear factor kappaB (NF-kappaB) by tumor necrosis factor alpha (TNFalpha). Here, we present evidence that TNFalpha induces the polyubiquitination of RIP1 at Lys-377 and that this polyubiquitination is required for the activation of IkappaB kinase (IKK) and NF-kappaB. A point mutation of RIP1 at Lys-377 (K377R) abolishes its polyubiquitination as well as its ability to restore IKK activation in a RIP1-deficient cell line. The K377R mutation of RIP1 also prevents the recruitment of TAK1 and IKK complexes to TNF receptor. Interestingly, polyubiquitinated RIP1 recruits IKK through the binding between the polyubiquitin chains and NEMO, a regulatory subunit of the IKK complex. Mutations of NEMO that disrupt its polyubiquitin binding also abolish IKK activation. These results reveal the biochemical mechanism underlying the essential signaling function of NEMO and provide direct evidence that signal-induced site-specific ubiquitination of RIP1 is required for IKK activation.


Assuntos
Proteínas de Transporte/metabolismo , Quinase I-kappa B/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poliubiquitina/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Sequência de Aminoácidos , Proteínas de Transporte/genética , Linhagem Celular , Ativação Enzimática , Humanos , Células Jurkat , Lisina/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Complexo de Proteínas Formadoras de Poros Nucleares/química , Complexo de Proteínas Formadoras de Poros Nucleares/deficiência , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Mutação Puntual , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Homologia de Sequência de Aminoácidos
18.
J Colloid Interface Sci ; 295(1): 124-34, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16168434

RESUMO

The modified Gouy-Chapman (MGC) theory has been used to study the electrical double layer near two charged plates immersed in a model electrolyte. The effects of assigning to the cations and anions different distances of closest approach to the charged surfaces are examined. The dependence of overcharging and charge reversal on the system parameters such as concentration, ion size and valence, is investigated both inside and outside the charged slit.

19.
Proc Natl Acad Sci U S A ; 102(49): 17717-22, 2005 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-16301520

RESUMO

Hepatitis C virus (HCV) is a global epidemic manifested mainly by chronic infection. One strategy that HCV employs to establish chronic infection is to use the viral Ser protease NS3/4A to cleave some unknown cellular targets involved in innate immunity. Here we show that the target of NS3/4A is the mitochondrial antiviral signaling protein, MAVS, that activates NF-kappaB and IFN regulatory factor 3 to induce type-I interferons. NS3/4A cleaves MAVS at Cys-508, resulting in the dislocation of the N-terminal fragment of MAVS from the mitochondria. Remarkably, a point mutation of MAVS at Cys-508 renders MAVS resistant to cleavage by NS3/4A, thus maintaining the ability of MAVS to induce interferons in HCV replicon cells. NS3/4A binds to and colocalizes with MAVS in the mitochondrial membrane, and it can cleave MAVS directly in vitro. These results provide an example of host-pathogen interaction in which the virus evades innate immunity by dislodging a pivotal antiviral protein from the mitochondria and suggest that blocking the cleavage of MAVS by NS3/4A may be applied to the prevention and treatment of HCV.


Assuntos
Proteínas de Transporte/metabolismo , Hepacivirus/enzimologia , Hepacivirus/imunologia , Imunidade Inata/imunologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas não Estruturais Virais/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/química , Linhagem Celular , Cisteína/genética , Cisteína/metabolismo , Hepacivirus/fisiologia , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Mitocôndrias/efeitos dos fármacos , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência , Transdução de Sinais/efeitos dos fármacos , Proteínas não Estruturais Virais/química , Proteínas Virais/química , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...