Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 360
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 18(1): 26-35, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27893701

RESUMO

TRAF1 is a signaling adaptor known for its role in tumor necrosis factor receptor-induced cell survival. Here we show that monocytes from healthy human subjects with a rheumatoid arthritis-associated single-nucleotide polymorphism (SNP) in the TRAF1 gene express less TRAF1 protein but greater amounts of inflammatory cytokines in response to lipopolysaccharide (LPS). The TRAF1 MATH domain binds directly to three components of the linear ubiquitination (LUBAC) complex, SHARPIN, HOIP and HOIL-1, to interfere with the recruitment and linear ubiquitination of NEMO. This results in decreased NF-κB activation and cytokine production, independently of tumor necrosis factor. Consistent with this, Traf1-/- mice show increased susceptibility to LPS-induced septic shock. These findings reveal an unexpected role for TRAF1 in negatively regulating Toll-like receptor signaling, providing a mechanistic explanation for the increased inflammation seen with a disease-associated TRAF1 SNP.


Assuntos
Artrite Reumatoide/genética , Leucócitos Mononucleares/imunologia , Monócitos/imunologia , Transdução de Sinais , Fator 1 Associado a Receptor de TNF/metabolismo , Animais , Citocinas/metabolismo , Predisposição Genética para Doença , Células HEK293 , Humanos , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polimorfismo de Nucleotídeo Único , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Fator 1 Associado a Receptor de TNF/genética , Receptores Toll-Like/metabolismo
2.
Cell ; 154(4): 859-74, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23953116

RESUMO

Mammalian target of rapamycin complex 1 (mTORC1) controls growth and survival in response to metabolic cues. Oxidative stress affects mTORC1 via inhibitory and stimulatory inputs. Whereas downregulation of TSC1-TSC2 activates mTORC1 upon oxidative stress, the molecular mechanism of mTORC1 inhibition remains unknown. Here, we identify astrin as an essential negative mTORC1 regulator in the cellular stress response. Upon stress, astrin inhibits mTORC1 association and recruits the mTORC1 component raptor to stress granules (SGs), thereby preventing mTORC1-hyperactivation-induced apoptosis. In turn, balanced mTORC1 activity enables expression of stress factors. By identifying astrin as a direct molecular link between mTORC1, SG assembly, and the stress response, we establish a unifying model of mTORC1 inhibition and activation upon stress. Importantly, we show that in cancer cells, apoptosis suppression during stress depends on astrin. Being frequently upregulated in tumors, astrin is a potential clinically relevant target to sensitize tumors to apoptosis.


Assuntos
Apoptose , Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Grânulos Citoplasmáticos/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Estresse Oxidativo , Proteína Regulatória Associada a mTOR
3.
Nat Immunol ; 15(11): 1079-89, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25282160

RESUMO

Humoral autoimmunity paralleled by the accumulation of follicular helper T cells (T(FH) cells) is linked to mutation of the gene encoding the RNA-binding protein roquin-1. Here we found that T cells lacking roquin caused pathology in the lung and accumulated as cells of the T(H)17 subset of helper T cells in the lungs. Roquin inhibited T(H)17 cell differentiation and acted together with the endoribonuclease regnase-1 to repress target mRNA encoding the T(H)17 cell-promoting factors IL-6, ICOS, c-Rel, IRF4, IκBNS and IκBζ. This cooperation required binding of RNA by roquin and the nuclease activity of regnase-1. Upon recognition of antigen by the T cell antigen receptor (TCR), roquin and regnase-1 proteins were cleaved by the paracaspase MALT1. Thus, this pathway acts as a 'rheostat' by translating TCR signal strength via graded inactivation of post-transcriptional repressors and differential derepression of targets to enhance T(H)17 differentiation.


Assuntos
Caspases/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Ribonucleases/metabolismo , Células Th17/citologia , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Genes rel/genética , Células HEK293 , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Fatores Reguladores de Interferon/genética , Interleucina-6/genética , Peptídeos e Proteínas de Sinalização Intracelular , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Knockout , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Proteínas Nucleares/genética , Proteínas/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Alinhamento de Sequência , Células Th17/imunologia , Ubiquitina-Proteína Ligases/genética
4.
EMBO J ; 40(13): e106777, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33999432

RESUMO

The p14ARF protein is a well-known regulator of p53-dependent and p53-independent tumor-suppressive activities. In unstressed cells, p14ARF is predominantly sequestered in the nucleoli, bound to its nucleolar interaction partner NPM. Upon genotoxic stress, p14ARF undergoes an immediate redistribution to the nucleo- and cytoplasm, where it promotes activation of cell cycle arrest and apoptosis. Here, we identify p14ARF as a novel interaction partner and substrate of PRMT1 (protein arginine methyltransferase 1). PRMT1 methylates several arginine residues in the C-terminal nuclear/nucleolar localization sequence (NLS/NoLS) of p14ARF . In the absence of cellular stress, these arginines are crucial for nucleolar localization of p14ARF . Genotoxic stress causes augmented interaction between PRMT1 and p14ARF , accompanied by arginine methylation of p14ARF . PRMT1-dependent NLS/NoLS methylation promotes the release of p14ARF from NPM and nucleolar sequestration, subsequently leading to p53-independent apoptosis. This PRMT1-p14ARF cooperation is cancer-relevant and indicative for PDAC (pancreatic ductal adenocarcinoma) prognosis and chemotherapy response of pancreatic tumor cells. Our data reveal that PRMT1-mediated arginine methylation is an important trigger for p14ARF 's stress-induced tumor-suppressive function.


Assuntos
Neoplasias Pancreáticas/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/patologia , Prognóstico , Células Sf9 , Proteína Supressora de Tumor p53/metabolismo , Neoplasias Pancreáticas
5.
Blood ; 141(9): 1023-1035, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35981498

RESUMO

Fms-like tyrosine kinase 3 (FLT3) is often overexpressed or constitutively activated by internal tandem duplication (ITD) and tyrosine kinase domain (TKD) mutations in acute myeloid leukemia (AML). Despite the use of receptor tyrosine kinase inhibitors (TKI) in FLT3-ITD-positive AML, the prognosis of patients is still poor, and further improvement of therapy is required. Targeting FLT3 independent of mutations by antibody-drug conjugates (ADCs) is a promising strategy for AML therapy. Here, we report the development and preclinical characterization of a novel FLT3-targeting ADC, 20D9-ADC, which was generated by applying the innovative P5 conjugation technology. In vitro, 20D9-ADC mediated potent cytotoxicity to Ba/F3 cells expressing transgenic FLT3 or FLT3-ITD, to AML cell lines, and to FLT3-ITD-positive patient-derived xenograft AML cells. In vivo, 20D9-ADC treatment led to a significant tumor reduction and even durable complete remission in AML xenograft models. Furthermore, 20D9-ADC demonstrated no severe hematotoxicity in in vitro colony formation assays using concentrations that were cytotoxic in AML cell line treatment. The combination of 20D9-ADC with the TKI midostaurin showed strong synergy in vitro and in vivo, leading to reduction of aggressive AML cells below the detection limit. Our data indicate that targeting FLT3 with an advanced new-generation ADC is a promising and potent antileukemic strategy, especially when combined with FLT3-TKI in FLT3-ITD-positive AML.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Tirosina Quinase 3 Semelhante a fms/genética , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Mutação
6.
Cell ; 136(3): 496-507, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19167051

RESUMO

Small regulatory RNAs including small interfering RNAs (siRNAs) and microRNAs (miRNAs) guide Argonaute (Ago) proteins to specific target RNAs leading to mRNA destabilization or translational repression. Here, we report the identification of Importin 8 (Imp8) as a component of miRNA-guided regulatory pathways. We show that Imp8 interacts with Ago proteins and localizes to cytoplasmic processing bodies (P bodies), structures involved in RNA metabolism. Furthermore, we detect Ago2 in the nucleus of HeLa cells, and knockdown of Imp8 reduces the nuclear Ago2 pool. Using immunoprecipitations of Ago2-associated mRNAs followed by microarray analysis, we further demonstrate that Imp8 is required for the recruitment of Ago protein complexes to a large set of Ago2-associated target mRNAs, allowing for efficient and specific gene silencing. Therefore, we provide evidence that Imp8 is required for cytoplasmic miRNA-guided gene silencing and affects nuclear localization of Ago proteins.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , RNA Mensageiro/metabolismo , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Proteínas Argonautas , Linhagem Celular , Grânulos Citoplasmáticos/metabolismo , Inativação Gênica , Células HeLa , Humanos , Corpos de Inclusão Intranuclear/metabolismo , MicroRNAs/metabolismo
7.
Development ; 147(21)2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32376681

RESUMO

Cilia are complex cellular protrusions consisting of hundreds of proteins. Defects in ciliary structure and function, many of which have not been characterised molecularly, cause ciliopathies: a heterogeneous group of human syndromes. Here, we report on the FOXJ1 target gene Cfap206, orthologues of which so far have only been studied in Chlamydomonas and Tetrahymena In mouse and Xenopus, Cfap206 was co-expressed with and dependent on Foxj1 CFAP206 protein localised to the basal body and to the axoneme of motile cilia. In Xenopus crispant larvae, the ciliary beat frequency of skin multiciliated cells was enhanced and bead transport across the epidermal mucociliary epithelium was reduced. Likewise, Cfap206 knockout mice revealed ciliary phenotypes. Electron tomography of immotile knockout mouse sperm flagella indicated a role in radial spoke formation reminiscent of FAP206 function in Tetrahymena Male infertility, hydrocephalus and impaired mucociliary clearance of the airways in the absence of laterality defects in Cfap206 mutant mice suggests that Cfap206 may represent a candidate for the subgroup of human primary ciliary dyskinesias caused by radial spoke defects.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Pulmão/metabolismo , Depuração Mucociliar , Motilidade dos Espermatozoides , Animais , Axonema/metabolismo , Corpos Basais/metabolismo , Cílios/metabolismo , Proteínas do Citoesqueleto/química , Desenvolvimento Embrionário , Células Epiteliais/metabolismo , Fluorescência , Hidrocefalia/patologia , Infertilidade Masculina/patologia , Masculino , Camundongos Knockout , Muco/metabolismo , Mutação/genética , Transporte Proteico , Espermatozoides/metabolismo , Espermatozoides/ultraestrutura , Xenopus laevis/embriologia , Xenopus laevis/metabolismo
8.
Nat Immunol ; 11(8): 725-33, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20639877

RESUMO

The molecular mechanism by which roquin controls the expression of inducible costimulator (ICOS) to prevent autoimmunity remains unsolved. Here we show that in helper T cells, roquin localized to processing (P) bodies and downregulated ICOS expression. The repression was dependent on the RNA helicase Rck, and roquin interacted with Rck and the enhancer of decapping Edc4, which act together in mRNA decapping. Sequences in roquin that confer P-body localization were essential for roquin-mediated ICOS repression. However, this process did not require microRNAs or the RNA-induced silencing complex (RISC). Instead, roquin bound ICOS mRNA directly, showing an intrinsic preference for a previously unrecognized sequence in the 3' untranslated region (3' UTR). Our results support a model in which roquin controls ICOS expression through binding to the 3' UTR of ICOS mRNA and by interacting with proteins that confer post-transcriptional repression.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , RNA Helicases DEAD-box/imunologia , MicroRNAs/genética , Proteínas Proto-Oncogênicas/imunologia , RNA Mensageiro/metabolismo , Transcrição Gênica , Ubiquitina-Proteína Ligases/metabolismo , Regiões 3' não Traduzidas , Sequência de Aminoácidos , Animais , Antígenos de Diferenciação de Linfócitos T/genética , Autoimunidade/genética , Autoimunidade/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica , Proteína Coestimuladora de Linfócitos T Induzíveis , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , MicroRNAs/imunologia , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Ubiquitina-Proteína Ligases/imunologia
9.
Immunity ; 38(4): 655-68, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23583643

RESUMO

The Roquin-1 protein binds to messenger RNAs (mRNAs) and regulates gene expression posttranscriptionally. A single point mutation in Roquin-1, but not gene ablation, increases follicular helper T (Tfh) cell numbers and causes lupus-like autoimmune disease in mice. In T cells, we did not identify a unique role for the much lower expressed paralog Roquin-2. However, combined ablation of both genes induced accumulation of T cells with an effector and follicular helper phenotype. We showed that Roquin-1 and Roquin-2 proteins redundantly repressed the mRNA of inducible costimulator (Icos) and identified the Ox40 costimulatory receptor as another shared mRNA target. Combined acute deletion increased Ox40 signaling, as well as Irf4 expression, and imposed Tfh differentiation on CD4(+) T cells. These data imply that both proteins maintain tolerance by preventing inappropriate T cell activation and Tfh cell differentiation, and that Roquin-2 compensates in the absence of Roquin-1, but not in the presence of its mutated form.


Assuntos
Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , RNA Mensageiro/metabolismo , Receptores OX40/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antígenos CD4/metabolismo , Diferenciação Celular/genética , Células HEK293 , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Ligação Proteica , Receptores OX40/genética , Proteínas Repressoras/genética , Ubiquitina-Proteína Ligases/genética
10.
Mol Cell ; 56(5): 630-40, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25454947

RESUMO

Proteolysis by aspartyl intramembrane proteases such as presenilin and signal peptide peptidase (SPP) underlies many cellular processes in health and disease. Saccharomyces cerevisiae encodes a homolog that we named yeast presenilin fold 1 (Ypf1), which we verify to be an SPP-type protease that localizes to the endoplasmic reticulum (ER). Our work shows that Ypf1 functionally interacts with the ER-associated degradation (ERAD) factors Dfm1 and Doa10 to regulate the abundance of nutrient transporters by degradation. We demonstrate how this noncanonical branch of the ERAD pathway, which we termed "ERAD regulatory" (ERAD-R), responds to ligand-mediated sensing as a trigger. More generally, we show that Ypf1-mediated posttranslational regulation of plasma membrane transporters is indispensible for early sensing and adaptation to nutrient depletion. The combination of systematic analysis alongside mechanistic details uncovers a broad role of intramembrane proteolysis in regulating secretome dynamics.


Assuntos
Retículo Endoplasmático/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Membrana Celular/metabolismo , Degradação Associada com o Retículo Endoplasmático , Regulação Fúngica da Expressão Gênica , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Filogenia , Saccharomyces cerevisiae/fisiologia , Alinhamento de Sequência , Ubiquitina-Proteína Ligases/metabolismo , Zinco/metabolismo
11.
Dev Biol ; 459(2): 109-125, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31884020

RESUMO

Malfunctions of motile cilia cause a variety of developmental defects and diseases in humans and animal model organisms. Defects include impaired mucociliary clearance of the airways, sperm immotility, hydrocephalus and organ laterality. Here, we characterize the evolutionary conserved Cfap43 gene by loss-of-function experiments in the mouse and the frog Xenopus laevis. Cfap43 is expressed in tissues carrying motile cilia and acts as a target gene of the transcription factor FOXJ1, which is essential for the induction of motile ciliogenesis. We show that CFAP43, a protein of unknown biochemical function, localizes to the ciliary axoneme. CFAP43 is involved in the regulation of the beating frequency of tracheal cilia and loss of CFAP43 causes severe mucus accumulation in the nasal cavity. Likewise, morphant and crispant frog embryos revealed impaired function of motile cilia of the larval epidermis, a model for airway mucociliary epithelia. CFAP43 participates in the formation of flagellar axonemes during spermatogenesis as mice mutant for Cfap43 display male infertility, consistent with observations in male sterile patients. In addition, mice mutant for Cfap43 display early onset hydrocephalus. Together, these results confirm the role of CFAP43 in the male reproductive tract and pinpoint additional functions in airway epithelia mucus clearance and brain development.


Assuntos
Cílios/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Proteínas do Citoesqueleto/genética , Células Epidérmicas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Hidrocefalia/genética , Infertilidade Masculina/genética , Masculino , Camundongos , Camundongos Knockout , Cauda do Espermatozoide/metabolismo , Espermatogênese/genética , Espermatozoides/metabolismo , Traqueia/citologia , Proteínas de Xenopus/genética , Xenopus laevis
12.
PLoS Pathog ; 15(5): e1007743, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31059555

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV; human herpesvirus 8) belongs to the subfamily of Gammaherpesvirinae and is the etiological agent of Kaposi's sarcoma as well as of two lymphoproliferative diseases: primary effusion lymphoma and multicentric Castleman disease. The KSHV life cycle is divided into a latent and a lytic phase and is highly regulated by viral immunomodulatory proteins which control the host antiviral immune response. Among them is a group of proteins with homology to cellular interferon regulatory factors, the viral interferon regulatory factors 1-4. The KSHV vIRFs are known as inhibitors of cellular interferon signaling and are involved in different oncogenic pathways. Here we characterized the role of the second vIRF protein, vIRF2, during the KSHV life cycle. We found the vIRF2 protein to be expressed in different KSHV positive cells with early lytic kinetics. Importantly, we observed that vIRF2 suppresses the expression of viral early lytic genes in both newly infected and reactivated persistently infected endothelial cells. This vIRF2-dependent regulation of the KSHV life cycle might involve the increased expression of cellular interferon-induced genes such as the IFIT proteins 1, 2 and 3, which antagonize the expression of early KSHV lytic proteins. Our findings suggest a model in which the viral protein vIRF2 allows KSHV to harness an IFN-dependent pathway to regulate KSHV early gene expression.


Assuntos
Endotélio Vascular/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interferons/metabolismo , Sarcoma de Kaposi/virologia , Proteínas Virais/metabolismo , Ativação Viral , Células Cultivadas , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Proteínas Imediatamente Precoces/genética , Fatores Reguladores de Interferon/genética , Interferons/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Proteínas Virais/genética , Latência Viral
13.
Nature ; 526(7573): 443-7, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26322584

RESUMO

Alzheimer disease (AD) is characterized by the accumulation of amyloid plaques, which are predominantly composed of amyloid-ß peptide. Two principal physiological pathways either prevent or promote amyloid-ß generation from its precursor, ß-amyloid precursor protein (APP), in a competitive manner. Although APP processing has been studied in great detail, unknown proteolytic events seem to hinder stoichiometric analyses of APP metabolism in vivo. Here we describe a new physiological APP processing pathway, which generates proteolytic fragments capable of inhibiting neuronal activity within the hippocampus. We identify higher molecular mass carboxy-terminal fragments (CTFs) of APP, termed CTF-η, in addition to the long-known CTF-α and CTF-ß fragments generated by the α- and ß-secretases ADAM10 (a disintegrin and metalloproteinase 10) and BACE1 (ß-site APP cleaving enzyme 1), respectively. CTF-η generation is mediated in part by membrane-bound matrix metalloproteinases such as MT5-MMP, referred to as η-secretase activity. η-Secretase cleavage occurs primarily at amino acids 504-505 of APP695, releasing a truncated ectodomain. After shedding of this ectodomain, CTF-η is further processed by ADAM10 and BACE1 to release long and short Aη peptides (termed Aη-α and Aη-ß). CTFs produced by η-secretase are enriched in dystrophic neurites in an AD mouse model and in human AD brains. Genetic and pharmacological inhibition of BACE1 activity results in robust accumulation of CTF-η and Aη-α. In mice treated with a potent BACE1 inhibitor, hippocampal long-term potentiation was reduced. Notably, when recombinant or synthetic Aη-α was applied on hippocampal slices ex vivo, long-term potentiation was lowered. Furthermore, in vivo single-cell two-photon calcium imaging showed that hippocampal neuronal activity was attenuated by Aη-α. These findings not only demonstrate a major functionally relevant APP processing pathway, but may also indicate potential translational relevance for therapeutic strategies targeting APP processing.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/citologia , Metaloproteinases da Matriz Associadas à Membrana/metabolismo , Neurônios/fisiologia , Proteólise , Proteínas ADAM/metabolismo , Proteína ADAM10 , Doença de Alzheimer/enzimologia , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/líquido cefalorraquidiano , Secretases da Proteína Precursora do Amiloide/deficiência , Secretases da Proteína Precursora do Amiloide/genética , Precursor de Proteína beta-Amiloide/líquido cefalorraquidiano , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/deficiência , Ácido Aspártico Endopeptidases/genética , Ácido Aspártico Endopeptidases/metabolismo , Sinalização do Cálcio , Modelos Animais de Doenças , Feminino , Hipocampo/enzimologia , Hipocampo/fisiologia , Humanos , Técnicas In Vitro , Potenciação de Longa Duração , Masculino , Metaloproteinases da Matriz Associadas à Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Peso Molecular , Neuritos/enzimologia , Neuritos/metabolismo , Neurônios/enzimologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Placa Amiloide , Processamento de Proteína Pós-Traducional , Análise de Célula Única
14.
Mol Cell ; 50(2): 236-49, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23562326

RESUMO

Centromere clustering during interphase is a phenomenon known to occur in many different organisms and cell types, yet neither the factors involved nor their physiological relevance is well understood. Using Drosophila tissue culture cells and flies, we identified a network of proteins, including the nucleoplasmin-like protein (NLP), the insulator protein CTCF, and the nucleolus protein Modulo, to be essential for the positioning of centromeres. Artificial targeting further demonstrated that NLP and CTCF are sufficient for clustering, while Modulo serves as the anchor to the nucleolus. Centromere clustering was found to depend on centric chromatin rather than specific DNA sequences. Moreover, unclustering of centromeres results in the spatial destabilization of pericentric heterochromatin organization, leading to partial defects in the silencing of repetitive elements, defects during chromosome segregation, and genome instability.


Assuntos
Nucléolo Celular/metabolismo , Centrômero/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Nucleoplasminas/metabolismo , Animais , Fator de Ligação a CCCTC , Linhagem Celular , Cromossomos de Insetos , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Técnicas de Silenciamento de Genes , Inativação Gênica , Instabilidade Genômica , Hemócitos/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Interfase , Nucleoplasminas/genética , Ligação Proteica , Mapas de Interação de Proteínas , Estabilidade Proteica , Transporte Proteico , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo
15.
Mol Cell ; 49(4): 668-79, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23317503

RESUMO

The HIV Nef protein recruits the polycomb protein Eed and mimics an integrin receptor signal for reasons that are not entirely clear. Here we demonstrate that Nef and Eed complex with the integrin effector paxillin to recruit and activate TNFα converting enzyme (TACE alias ADAM 17) and its close relative ADAM10. The activated proteases cleaved proTNFα and were shuttled into extracellular vesicles (EVs). Peripheral blood mononuclear cells that ingested these EVs released TNFα. Analyzing the mechanism, we found that Pak2, an established host cell effector of Nef, phosphorylated paxillin on Ser272/274 to induce TACE-paxillin association and shuttling into EVs via lipid rafts. Conversely, Pak1 phosphorylated paxillin on Ser258, which inhibited TACE association and lipid raft transfer. Interestingly, melanoma cells used an identical mechanism to shuttle predominantly ADAM10 into EVs. We conclude that HIV-1 and cancer cells exploit a paxillin/integrin-controlled mechanism to release TACE/ADAM10-containing vesicles, ensuring better proliferation/growth conditions in their microenvironment.


Assuntos
Proteínas ADAM/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Proteínas de Membrana/metabolismo , Paxilina/fisiologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/fisiologia , Quinases Ativadas por p21/fisiologia , Proteínas ADAM/sangue , Proteína ADAM10 , Proteína ADAM17 , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Substituição de Aminoácidos , Secretases da Proteína Precursora do Amiloide/sangue , Estudos de Casos e Controles , Ativação Enzimática , Células HEK293 , Infecções por HIV/sangue , Infecções por HIV/enzimologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo K , Humanos , Melanoma/sangue , Melanoma/enzimologia , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/sangue , Mutagênese Sítio-Dirigida , Paxilina/genética , Paxilina/metabolismo , Fosforilação , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica , Proteína Quinase C-delta/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Ribonucleoproteínas/metabolismo , Vesículas Secretórias/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Quinases Ativadas por p21/metabolismo
16.
Nucleic Acids Res ; 47(14): 7444-7459, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31147711

RESUMO

Preblastoderm Drosophila embryo development is characterized by fast cycles of nuclear divisions. Extracts from these embryos can be used to reconstitute complex chromatin with high efficiency. We now discovered that this chromatin assembly system contains activities that recognize unprotected DNA ends and signal DNA damage through phosphorylation. DNA ends are initially bound by Ku and MRN complexes. Within minutes, the phosphorylation of H2A.V (homologous to γH2A.X) initiates from DNA breaks and spreads over tens of thousands DNA base pairs. The γH2A.V phosphorylation remains tightly associated with the damaged DNA and does not spread to undamaged DNA in the same reaction. This first observation of long-range γH2A.X spreading along damaged chromatin in an in vitro system provides a unique opportunity for mechanistic dissection. Upon further incubation, DNA ends are rendered single-stranded and bound by the RPA complex. Phosphoproteome analyses reveal damage-dependent phosphorylation of numerous DNA-end-associated proteins including Ku70, RPA2, CHRAC16, the exonuclease Rrp1 and the telomer capping complex. Phosphorylation of spindle assembly checkpoint components and of microtubule-associated proteins required for centrosome integrity suggests this cell-free system recapitulates processes involved in the regulated elimination of fatally damaged syncytial nuclei.


Assuntos
Sistema Livre de Células/metabolismo , Quebras de DNA , Drosophila/genética , Transdução de Sinais , Animais , Linhagem Celular , Cromatina/genética , Cromatina/metabolismo , Reparo do DNA , Drosophila/citologia , Drosophila/embriologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Histonas/genética , Histonas/metabolismo , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Fosforilação , Proteoma/genética , Proteoma/metabolismo , Proteômica/métodos
17.
Int J Cancer ; 146(5): 1396-1408, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31525266

RESUMO

Chitinase-like proteins (CLP) are chitin-binding proteins that lack chitin hydrolyzing activity, but possess cytokine-like and growth factor-like properties, and play crucial role in intercellular crosstalk. Both human and mice express two members of CLP family: YKL-40 and stabilin-1 interacting chitinase-like protein (SI-CLP). Despite numerous reports indicating the role of YKL-40 in the support of angiogenesis, tumor cell proliferation, invasion and metastasis, the role of its structurally related protein SI-CLP in cancer was not reported. Using gain-of-function approach, we demonstrate in the current study that the expression of recombinant SI-CLP in mouse TS/A mammary adenocarcinoma cells results in significant and persistent inhibition of in vivo tumor growth. Using quantitative immunohistochemistry, we show that on the cellular level this phenomenon is associated with reduced infiltration of tumor-associated macrophages (TAMs), CD4+ and FoxP3+ cells in SI-CLP expressing tumors. Gene expression analysis in TAM isolated from SI-CLP-expressing and control tumors demonstrated that SI-CLP does not affect macrophage phenotype. However, SI-CLP significantly inhibited migration of murine bone-marrow derived macrophages and human primary monocytes toward monocyte-recruiting chemokine CCL2 produced in the tumor microenvironment (TME). Mechanistically, SI-CLP did not affect CCL2/CCR2 interaction, but suppressed cytoskeletal rearrangements in response to CCL2. Altogether, our data indicate that SI-CLP functions as a tumor growth inhibitor in mouse breast cancer by altering cellular composition of TME and blocking cytokine-induced TAM recruitment. Taking into consideration weak to absent expression of SI-CLP in human breast cancer, it can be considered as a therapeutic protein to block TAM-mediated support of breast tumor growth.


Assuntos
Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Transporte/imunologia , Macrófagos/imunologia , Neoplasias Mamárias Experimentais/imunologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Processos de Crescimento Celular/imunologia , Movimento Celular/imunologia , Feminino , Células HEK293 , Humanos , Ativação de Macrófagos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade
18.
Immunity ; 35(6): 945-57, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22195748

RESUMO

Little is known about mechanisms determining the homeostasis of lymphocytes within lymphoid organs. Applying different mouse models, including conditionally proficient Ccr7 gene-targeted mice, we now show that semimature steady state dendritic cells (sDCs) constitutively trafficking into lymph nodes (LNs) were essential contributors to T cell homeostasis in these organs. sDCs provided vascular endothelial growth factor known to support high endothelial venule formation, thus facilitating enhanced homing of T cells to LNs. The presence of sDCs led to increased CCL21 production in T-zone fibroblastic reticular cells. CCL21 is a ligand for CCR7 known to regulate homing as well as retention of T cells in LNs. In addition, we provide evidence that CCL21 binds to the surface of DCs via its heparin-binding domain, further explaining why T cells leave LNs more rapidly in the absence of sDCs. Together, these data reveal multiple roles for sDCs in regulating T cell homeostasis in LNs.


Assuntos
Células Dendríticas/imunologia , Linfonodos/imunologia , Linfócitos T/imunologia , Animais , Células da Medula Óssea/metabolismo , Movimento Celular/imunologia , Quimiocina CCL21/metabolismo , Quimerismo , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células Dendríticas/metabolismo , Marcação de Genes , Homeostase/genética , Homeostase/imunologia , Humanos , Linfonodos/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Receptores CCR7/genética , Receptores CCR7/imunologia , Células Estromais/metabolismo , Linfócitos T/metabolismo
19.
J Immunol ; 200(2): 558-564, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29222166

RESUMO

IL-7 therapy has been evaluated in patients who do not regain normal CD4 T cell counts after virologically successful antiretroviral therapy. IL-7 increases total circulating CD4 and CD8 T cell counts; however, its effect on HIV-specific CD8 T cells has not been fully examined. TRAF1, a prosurvival signaling adaptor required for 4-1BB-mediated costimulation, is lost from chronically stimulated virus-specific CD8 T cells with progression of HIV infection in humans and during chronic lymphocytic choriomeningitis infection in mice. Previous results showed that IL-7 can restore TRAF1 expression in virus-specific CD8 T cells in mice, rendering them sensitive to anti-4-1BB agonist therapy. In this article, we show that IL-7 therapy in humans increases the number of circulating HIV-specific CD8 T cells. For a subset of patients, we also observed an increased frequency of TRAF1+ HIV-specific CD8 T cells 10 wk after completion of IL-7 treatment. IL-7 treatment increased levels of phospho-ribosomal protein S6 in HIV-specific CD8 T cells, suggesting increased activation of the metabolic checkpoint kinase mTORC1. Thus, IL-7 therapy in antiretroviral therapy-treated patients induces sustained changes in the number and phenotype of HIV-specific T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , HIV-1/imunologia , Proteína S6 Ribossômica/metabolismo , Fator 1 Associado a Receptor de TNF/metabolismo , Terapia Antirretroviral de Alta Atividade , Contagem de Linfócito CD4 , Citocinas/biossíntese , Expressão Gênica , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Interleucina-7/farmacologia , Interleucina-7/uso terapêutico , Contagem de Linfócitos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Ligação Proteica , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Proteína S6 Ribossômica/genética , Fator 1 Associado a Receptor de TNF/genética , Carga Viral
20.
Mol Cell ; 48(3): 434-44, 2012 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-23022379

RESUMO

Packaging of DNA into nucleosomes and the formation of higher-order chromatin structures determine DNA accessibility and activity of genome domains. We identified an RNA-dependent mechanism maintaining the open chromatin structure within euchromatic regions in Drosophila cells. The mechanism of reversible chromatin opening, reconstituted in vitro, depends on the Drosophila decondensation factor 31 (Df31) that specifically binds to RNA and localizes to euchromatic regions. Df31 is capable to tether a heterogeneous pool of short, single-stranded RNAs to chromatin. This class of chromatin-associated RNA (caRNA) is stably linked to chromatin and is largely composed of snoRNAs, which are preferentially bound by Df31. We suggest that the Df31-mediated linkage of snoRNAs and chromatin, forms a RNA-chromatin network resulting in the establishment of open chromatin domains. Analysis of caRNAs in human cells also reveals a strong enrichment of snoRNAs, implying a conserved role for these molecules in higher-order structures of chromatin.


Assuntos
Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Proteínas de Drosophila/genética , RNA Nucleolar Pequeno/genética , Animais , Sequência de Bases , Linhagem Celular , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Células HeLa , Histonas/metabolismo , Humanos , Imunoprecipitação , Dados de Sequência Molecular , Nucleossomos/genética , Nucleossomos/metabolismo , Ligação Proteica , RNA Nucleolar Pequeno/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA