Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 57(1): 39-54, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25498145

RESUMO

The lysosome is the final destination for degradation of endocytic cargo, plasma membrane constituents, and intracellular components sequestered by macroautophagy. Fusion of endosomes and autophagosomes with the lysosome depends on the GTPase Rab7 and the homotypic fusion and protein sorting (HOPS) complex, but adaptor proteins that link endocytic and autophagy pathways with lysosomes are poorly characterized. Herein, we show that Pleckstrin homology domain containing protein family member 1 (PLEKHM1) directly interacts with HOPS complex and contains a LC3-interacting region (LIR) that mediates its binding to autophagosomal membranes. Depletion of PLEKHM1 blocks lysosomal degradation of endocytic (EGFR) cargo and enhances presentation of MHC class I molecules. Moreover, genetic loss of PLEKHM1 impedes autophagy flux upon mTOR inhibition and PLEKHM1 regulates clearance of protein aggregates in an autophagy- and LIR-dependent manner. PLEKHM1 is thus a multivalent endocytic adaptor involved in the lysosome fusion events controlling selective and nonselective autophagy pathways.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Lisossomos/metabolismo , Fusão de Membrana/genética , Glicoproteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Fagossomos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Proteínas Reguladoras de Apoptose , Autofagia , Proteínas Relacionadas à Autofagia , Endossomos/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Transdução de Sinais , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
2.
Haematologica ; 107(4): 803-815, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33910331

RESUMO

Although great advances have been made in understanding the pathobiology of mixed lineage leukemia-rearranged (MLL-r) leukemias, therapies for this leukemia have remained limited, and clinical outcomes remain bleak. In order to identify novel targets for immunotherapy treatments, we compiled a lineage-independent MLL-r leukemia gene signature using publicly available data sets. Data from large leukemia repositories were filtered through the in silico human surfaceome, providing a list of highly predicted cell surface proteins overexpressed in MLL-r leukemias. LAMP5, a lysosomal associated membrane protein, is expressed highly and specifically in MLL-r leukemia. We found that LAMP5 is a direct target of the oncogenic MLL-fusion protein. LAMP5 depletion significantly inhibited leukemia cell growth in vitro and in vivo. Functional studies showed that LAMP-5 is a novel modulator of innate-immune pathways in MLL-r leukemias. Downregulation of LAMP5 led to inhibition of NF-kB signaling and increased activation of type-1 interferon signaling downstream of Toll-like receptor/interleukin 1 receptor activation. These effects were attributable to the critical role of LAMP-5 in transferring the signal flux from interferon signaling endosomes to pro-inflammatory signaling endosomes. Depletion of IRF7 was able to partially rescue the cell growth inhibition upon LAMP5 downregulation. Lastly, LAMP-5 was readily detected on the surface of MLL-r leukemia cells. Targeting surface LAMP-5 using an antibody-drug conjugate leads to significant cell viability decrease specifically in MLL-r leukemias. Overall, based on the limited expression throughout human tissues, we postulate that LAMP-5 could potentially serve as an immunotherapeutic target with a wide therapeutic window to treat MLL-r leukemias.


Assuntos
Leucemia Aguda Bifenotípica , Leucemia , Histona-Lisina N-Metiltransferase/genética , Humanos , Imunoterapia , Leucemia/genética , Proteína de Leucina Linfoide-Mieloide/genética , Proteína de Leucina Linfoide-Mieloide/metabolismo
3.
Proc Natl Acad Sci U S A ; 116(45): 22721-22729, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31636192

RESUMO

Exposure to microbe-associated molecular patterns (MAMPs) causes dendritic cells (DCs) to undergo a remarkable activation process characterized by changes in key biochemical mechanisms. These enhance antigen processing and presentation, as well as strengthen DC capacity to stimulate naïve T cell proliferation. Here, we show that in response to the MAMPS lipopolysaccharide and polyriboinosinic:polyribocytidylic acid (Poly I:C), RNA polymerase III (Pol lII)-dependent transcription and consequently tRNA gene expression are strongly induced in DCs. This is in part caused by the phosphorylation and nuclear export of MAF1 homolog negative regulator of Poll III (MAF1), via a synergistic casein kinase 2 (CK2)- and mammalian target of rapamycin-dependent signaling cascade downstream of Toll-like receptors (TLRs). De novo tRNA expression is necessary to augment protein synthesis and compensate for tRNA degradation driven by TLR-dependent DC exposure to type-I IFN. Although protein synthesis is not strongly inhibited in absence of RNA Pol III activity, it compromises the translation of key DC mRNAs, like those coding for costimulatory molecules and proinflammatory cytokines, which instead can be stored in stress granules, as shown for CD86 mRNA. TLR-dependent CK2 stimulation and subsequent RNA Pol III activation are therefore key for the acquisition by DCs of their unique T cell immune-stimulatory functions.


Assuntos
Células Dendríticas/imunologia , RNA Polimerase III/genética , Linfócitos T/imunologia , Transcrição Gênica , Animais , Caseína Quinase II/metabolismo , Células Cultivadas , Ativação Enzimática , Feminino , Camundongos , Fosforilação , RNA Polimerase III/metabolismo , RNA de Transferência/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo
4.
EMBO J ; 36(6): 761-782, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28100675

RESUMO

In innate immune responses, induction of type-I interferons (IFNs) prevents virus spreading while viral replication is delayed by protein synthesis inhibition. We asked how cells perform these apparently contradictory activities. Using single fibroblast monitoring by flow cytometry and mathematical modeling, we demonstrate that type-I IFN production is linked to cell's ability to enter dsRNA-activated PKR-dependent translational arrest and then overcome this inhibition by decreasing eIF2α phosphorylation through phosphatase 1c cofactor GADD34 (Ppp1r15a) expression. GADD34 expression, shown here to be dependent on the IRF3 transcription factor, is responsible for a biochemical cycle permitting pulse of IFN synthesis to occur in cells undergoing protein synthesis inhibition. Translation arrest is further demonstrated to be key for anti-viral response by acting synergistically with MAVS activation to amplify TBK1 signaling and IFN-ß mRNA transcription, while GADD34-dependent protein synthesis recovery contributes to the heterogeneous expression of IFN observed in dsRNA-activated cells.


Assuntos
Regulação da Expressão Gênica , Interferon beta/metabolismo , Biossíntese de Proteínas , Proteína Fosfatase 1/metabolismo , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , Animais , Células Cultivadas , Fibroblastos/imunologia , Fibroblastos/virologia , Citometria de Fluxo , Perfilação da Expressão Gênica , Imunidade Inata , Camundongos , Modelos Teóricos
5.
J Cell Sci ; 131(10)2018 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-29700204

RESUMO

The rate at which ribosomes translate mRNAs regulates protein expression by controlling co-translational protein folding and mRNA stability. Many factors regulate translation elongation, including tRNA levels, codon usage and phosphorylation of eukaryotic elongation factor 2 (eEF2). Current methods to measure translation elongation lack single-cell resolution, require expression of multiple transgenes and have never been successfully applied ex vivo Here, we show, by using a combination of puromycilation detection and flow cytometry (a method we call 'SunRiSE'), that translation elongation can be measured accurately in primary cells in pure or heterogenous populations isolated from blood or tissues. This method allows for the simultaneous monitoring of multiple parameters, such as mTOR or S6K1/2 signaling activity, the cell cycle stage and phosphorylation of translation factors in single cells, without elaborated, costly and lengthy purification procedures. We took advantage of SunRiSE to demonstrate that, in mouse embryonic fibroblasts, eEF2 phosphorylation by eEF2 kinase (eEF2K) mostly affects translation engagement, but has a surprisingly small effect on elongation, except after proteotoxic stress induction.This article has an associated First Person interview with the first author of the paper.


Assuntos
Fibroblastos/citologia , Citometria de Fluxo/métodos , Elongação Traducional da Cadeia Peptídica , Análise de Célula Única/métodos , Animais , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Fibroblastos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Biossíntese de Proteínas , Proteínas/genética , Proteínas/metabolismo , Ribossomos/genética , Ribossomos/metabolismo
7.
Immunol Rev ; 272(1): 28-38, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27319340

RESUMO

Antigenic peptides presented in the context of major histocompatibility complex (MHC) molecules originate from the degradation of both self and non-self proteins. T cells can therefore recognize at the surface of surveyed cells, the self-peptidome produced by the cell itself (mostly inducing tolerance) or immunogenic peptides derived from exogenous origins. The initiation of adaptive immune responses by dendritic cells (DCs), through the antigenic priming of naïve T cells, is associated to microbial pattern recognition receptors engagement. Activation of DCs by microbial product or inflammatory cytokines initiates multiple processes that maximize DC capacity to present exogenous antigens and stimulate T cells by affecting major metabolic and membrane traffic pathways. These include the modulation of protein synthesis, the regulation of MHC and co-stimulatory molecules transport, as well as the regulation of autophagy, that, all together promote exogenous antigen presentation while limiting the display of self-antigens by MHC molecules.


Assuntos
Apresentação de Antígeno , Autofagia , Células Dendríticas/imunologia , Biossíntese de Proteínas , Linfócitos T/imunologia , Imunidade Adaptativa , Animais , Autoantígenos/metabolismo , Diferenciação Celular , Antígenos de Histocompatibilidade/metabolismo , Humanos , Imunomodulação , Peptídeos/metabolismo , Tolerância a Antígenos Próprios
8.
Biochemistry ; 56(31): 4029-4038, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28703578

RESUMO

Transfer RNAs (tRNAs) are among the most heavily modified RNA species. Posttranscriptional tRNA modifications (ptRMs) play fundamental roles in modulating tRNA structure and function and are being increasingly linked to human physiology and disease. Detection of ptRMs is often challenging, expensive, and laborious. Restriction fragment length polymorphism (RFLP) analyses study the patterns of DNA cleavage after restriction enzyme treatment and have been used for the qualitative detection of modified bases on mRNAs. It is known that some ptRMs induce specific and reproducible base "mutations" when tRNAs are reverse transcribed. For example, inosine, which derives from the deamination of adenosine, is detected as a guanosine when an inosine-containing tRNA is reverse transcribed, amplified via polymerase chain reaction (PCR), and sequenced. ptRM-dependent base changes on reverse transcription PCR amplicons generated as a consequence of the reverse transcription reaction might create or abolish endonuclease restriction sites. The suitability of RFLP for the detection and/or quantification of ptRMs has not been studied thus far. Here we show that different ptRMs can be detected at specific sites of different tRNA types by RFLP. For the examples studied, we show that this approach can reliably estimate the modification status of the sample, a feature that can be useful in the study of the regulatory role of tRNA modifications in gene expression.


Assuntos
Adenosina Desaminase/metabolismo , Modelos Biológicos , Polimorfismo de Fragmento de Restrição , Processamento Pós-Transcricional do RNA , RNA de Transferência de Alanina/metabolismo , RNA de Transferência de Treonina/metabolismo , Adenosina/metabolismo , Adenosina Desaminase/química , Adenosina Desaminase/genética , Análise do Polimorfismo de Comprimento de Fragmentos Amplificados , Pareamento de Bases , Biologia Computacional , Desaminação , Sistemas Inteligentes , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Inosina/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA de Transferência de Alanina/antagonistas & inibidores , RNA de Transferência de Treonina/antagonistas & inibidores , RNA de Transferência de Valina/antagonistas & inibidores , RNA de Transferência de Valina/metabolismo , Transcrição Reversa , Especificidade por Substrato
9.
EMBO J ; 32(9): 1214-24, 2013 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-23584529

RESUMO

The innate immune cell network detects specific microbes and damages to cell integrity in order to coordinate and polarize the immune response against invading pathogens. In recent years, a cross-talk between microbial-sensing pathways and endoplasmic reticulum (ER) homeostasis has been discovered and have attracted the attention of many researchers from the inflammation field. Abnormal accumulation of proteins in the ER can be seen as a sign of cellular malfunction and triggers a collection of conserved emergency rescue pathways. These signalling cascades, which increase ER homeostasis and favour cell survival, are collectively known as the unfolded protein response (UPR). The induction or activation by microbial stimuli of several molecules linked to the ER stress response pathway have led to the conclusion that microbe sensing by immunocytes is generally associated with an UPR, which serves as a signal amplification cascade favouring inflammatory cytokines production. Induction of the UPR alone was shown to promote inflammation in different cellular and pathological models. Here we discuss how the innate immune and ER-signalling pathways intersect. Moreover, we propose that the induction of UPR-related molecules by microbial products does not necessarily reflect ER stress, but instead is an integral part of a specific transcription programme controlled by innate immunity receptors.


Assuntos
Mapeamento Cromossômico , Imunidade Ativa/genética , Receptor Cross-Talk/fisiologia , Transdução de Sinais/genética , Estresse Fisiológico/genética , Animais , Estresse do Retículo Endoplasmático/genética , Estresse do Retículo Endoplasmático/imunologia , Humanos , Imunidade Ativa/imunologia , Modelos Biológicos , Receptor Cross-Talk/imunologia , Transdução de Sinais/imunologia , Estresse Fisiológico/imunologia
10.
Immunol Cell Biol ; 95(9): 753-764, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28559542

RESUMO

Given the heterogeneous nature of antigens, major histocompatibility complex class I (MHC I) intracellular transport intersects with multiple degradation pathways for efficient peptide loading and presentation to cytotoxic T cells. MHC I loading with peptides in the endoplasmic reticulum (ER) is a tightly regulated process, while post-ER intracellular transport is considered to occur by default, leading to peptide-bearing MHC I delivery to the plasma membrane. We show here that MHC I traffic is submitted to a previously uncharacterized sorting step at the trans Golgi network (TGN), dependent on the ubiquitination of its cytoplasmic tail lysine residues. MHC I ubiquitination is mediated by the E3 ligase membrane-associated RING-CH 9 (MARCH9) and allows MHC I access to Syntaxin 6-positive endosomal compartments. We further show that MARCH9 can also target the human MHC I-like lipid antigen-presentation molecule CD1a. MARCH9 expression is modulated by microbial pattern exposure in dendritic cells (DCs), thus revealing the role of this ubiquitin E3 ligase in coordinating MHC I access to endosomes and DC activation for efficient antigen cross-presentation.


Assuntos
Antígenos CD1/metabolismo , Membrana Celular/metabolismo , Células Dendríticas/imunologia , Endossomos/metabolismo , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Rede trans-Golgi/metabolismo , Apresentação de Antígeno , Antígenos CD1/genética , Células Cultivadas , Retículo Endoplasmático/metabolismo , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Proteínas de Membrana , Monócitos/imunologia , Domínios Proteicos/genética , Sinais Direcionadores de Proteínas/genética , Transporte Proteico , Proteínas Qa-SNARE/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
11.
Proc Natl Acad Sci U S A ; 109(8): 3006-11, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22315398

RESUMO

In response to inflammatory stimulation, dendritic cells (DCs) have a remarkable pattern of differentiation that exhibits specific mechanisms to control the immune response. Here we show that in response to polyriboinosinic:polyribocytidylic acid (pI:C), DCs mount a specific integrated stress response during which the transcription factor ATF4 and the growth arrest and DNA damage-inducible protein 34 (GADD34/Ppp1r15a), a phosphatase 1 (PP1) cofactor, are expressed. In agreement with increased GADD34 levels, an extensive dephosphorylation of the translation initiation factor eIF2α was observed during DC activation. Unexpectedly, although DCs display an unusual resistance to protein synthesis inhibition induced in response to cytosolic dsRNA, GADD34 expression did not have a major impact on protein synthesis. GADD34, however, was shown to be required for normal cytokine production both in vitro and in vivo. These observations have important implications in linking further pathogen detection with the integrated stress response pathways.


Assuntos
Citocinas/biossíntese , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/enzimologia , Poli I-C/farmacologia , Proteína Fosfatase 1/metabolismo , Subunidades Proteicas/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Animais , Citosol/efeitos dos fármacos , Citosol/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fator de Iniciação 2 em Eucariotos/metabolismo , Interferon beta/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , eIF-2 Quinase/metabolismo
12.
PLoS Pathog ; 8(5): e1002708, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615568

RESUMO

Nucleic acid sensing by cells is a key feature of antiviral responses, which generally result in type-I Interferon production and tissue protection. However, detection of double-stranded RNAs in virus-infected cells promotes two concomitant and apparently conflicting events. The dsRNA-dependent protein kinase (PKR) phosphorylates translation initiation factor 2-alpha (eIF2α) and inhibits protein synthesis, whereas cytosolic DExD/H box RNA helicases induce expression of type I-IFN and other cytokines. We demonstrate that the phosphatase-1 cofactor, growth arrest and DNA damage-inducible protein 34 (GADD34/Ppp1r15a), an important component of the unfolded protein response (UPR), is absolutely required for type I-IFN and IL-6 production by mouse embryonic fibroblasts (MEFs) in response to dsRNA. GADD34 expression in MEFs is dependent on PKR activation, linking cytosolic microbial sensing with the ATF4 branch of the UPR. The importance of this link for anti-viral immunity is underlined by the extreme susceptibility of GADD34-deficient fibroblasts and neonate mice to Chikungunya virus infection.


Assuntos
Infecções por Alphavirus/imunologia , Vírus Chikungunya/imunologia , Interferon beta/biossíntese , Proteína Fosfatase 1/metabolismo , RNA de Cadeia Dupla/imunologia , Células 3T3 , Fator 4 Ativador da Transcrição/metabolismo , Animais , Linhagem Celular , Febre de Chikungunya , Fibroblastos/imunologia , Fibroblastos/virologia , Interferon Tipo I/biossíntese , Interleucina-6/biossíntese , Camundongos , Poli I-C/imunologia , Proteína Fosfatase 1/biossíntese , Proteína Fosfatase 1/genética , Tapsigargina/imunologia , Resposta a Proteínas não Dobradas , eIF-2 Quinase/biossíntese , eIF-2 Quinase/metabolismo
13.
RNA Biol ; 11(9): 1199-213, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25483040

RESUMO

Mutations in genes that encode tRNAs, aminoacyl-tRNA syntheases, tRNA modifying enzymes and other tRNA interacting partners are associated with neuropathies, cancer, type-II diabetes and hearing loss, but how these mutations cause disease is unclear. We have hypothesized that levels of tRNA decoding error (mistranslation) that do not fully impair embryonic development can accelerate cell degeneration through proteome instability and saturation of the proteostasis network. To test this hypothesis we have induced mistranslation in zebrafish embryos using mutant tRNAs that misincorporate Serine (Ser) at various non-cognate codon sites. Embryo viability was affected and malformations were observed, but a significant proportion of embryos survived by activating the unfolded protein response (UPR), the ubiquitin proteasome pathway (UPP) and downregulating protein biosynthesis. Accumulation of reactive oxygen species (ROS), mitochondrial and nuclear DNA damage and disruption of the mitochondrial network, were also observed, suggesting that mistranslation had a strong negative impact on protein synthesis rate, ER and mitochondrial homeostasis. We postulate that mistranslation promotes gradual cellular degeneration and disease through protein aggregation, mitochondrial dysfunction and genome instability.


Assuntos
Códon/genética , Embrião não Mamífero/citologia , Mutação/genética , Biossíntese de Proteínas , Proteínas/metabolismo , RNA de Transferência/genética , Peixe-Zebra/genética , Animais , Northern Blotting , Western Blotting , Núcleo Celular/genética , Dano ao DNA/genética , DNA Mitocondrial/genética , Embrião não Mamífero/fisiologia , Retículo Endoplasmático/metabolismo , Estresse Oxidativo , Complexo de Endopeptidases do Proteassoma/genética , Processamento de Proteína Pós-Traducional , Proteoma/análise , Espécies Reativas de Oxigênio/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Peixe-Zebra/embriologia
14.
Blood ; 118(3): 609-17, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21642595

RESUMO

The brain and dendritic cell (BAD)-associated lysosome-associated membrane protein (LAMP)-like molecule (BAD-LAMP, c20orf103, UNC-46) is a newly identified member of the family of LAMPs. BAD-LAMP expression in the mouse is confined to neurons. We demonstrate here that in humans, BAD-LAMP can specifically be found in the type I IFN-producing plasmacytoid dendritic cells (pDCs). Human BAD-LAMP is localized in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) of freshly isolated CD123(+) pDCs and is rapidly lost upon activation by unmethylated cytosine-phosphate-guanine (CpG) oligonucleotides. The restricted pattern of BAD-LAMP expression allows for the rapid identification of normal and leukemic human pDCs in tissues and blood.


Assuntos
Biomarcadores/metabolismo , Células Dendríticas/metabolismo , Leucemia/patologia , Proteínas de Membrana Lisossomal/metabolismo , Proteínas de Membrana/metabolismo , Encéfalo/metabolismo , Células Dendríticas/citologia , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Interleucina-3/metabolismo , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Leucemia/metabolismo , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Transfecção
15.
Nat Commun ; 14(1): 4290, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37463962

RESUMO

Endo-lysosomes transport along microtubules and clustering in the perinuclear area are two necessary steps for microbes to activate specialized phagocyte functions. We report that RUN and FYVE domain-containing protein 3 (RUFY3) exists as two alternative isoforms distinguishable by the presence of a C-terminal FYVE domain and by their affinity for phosphatidylinositol 3-phosphate on endosomal membranes. The FYVE domain-bearing isoform (iRUFY3) is preferentially expressed in primary immune cells and up-regulated upon activation by microbes and Interferons. iRUFY3 is necessary for ARL8b + /LAMP1+ endo-lysosomes positioning in the pericentriolar organelles cloud of LPS-activated macrophages. We show that iRUFY3 controls macrophages migration, MHC II presentation and responses to Interferon-γ, while being important for intracellular Salmonella replication. Specific inactivation of rufy3 in phagocytes leads to aggravated pathologies in mouse upon LPS injection or bacterial pneumonia. This study highlights the role of iRUFY3 in controlling endo-lysosomal dynamics, which contributes to phagocyte activation and immune response regulation.


Assuntos
Apresentação de Antígeno , Lipopolissacarídeos , Animais , Camundongos , Endossomos/metabolismo , Lipopolissacarídeos/metabolismo , Lisossomos/metabolismo , Fagócitos
16.
J Virol ; 85(1): 606-20, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20962078

RESUMO

Chikungunya virus (CHIKV) is an arthritogenic mosquito-transmitted alphavirus that is undergoing reemergence in areas around the Indian Ocean. Despite the current and potential danger posed by this virus, we know surprisingly little about the induction and evasion of CHIKV-associated antiviral immune responses. With this in mind we investigated innate immune reactions to CHIKV in human fibroblasts, a demonstrable in vivo target of virus replication and spread. We show that CHIKV infection leads to activation of the transcription factor interferon regulatory factor 3 (IRF3) and subsequent transcription of IRF3-dependent antiviral genes, including beta interferon (IFN-ß). IRF3 activation occurs by way of a virus-induced innate immune signaling pathway that includes the adaptor molecule interferon promoter stimulator 1 (IPS-1). Despite strong transcriptional upregulation of these genes, however, translation of the corresponding proteins is not observed. We further demonstrate that translation of cellular (but not viral) genes is blocked during infection and that although CHIKV is found to trigger inactivation of the translational molecule eukaryotic initiation factor subunit 2α by way of the double-stranded RNA sensor protein kinase R, this response is not required for the block to protein synthesis. Furthermore, overall diminution of cellular RNA synthesis is also observed in the presence of CHIKV and transcription of IRF3-dependent antiviral genes appears specifically blocked late in infection. We hypothesize that the observed absence of IFN-ß and antiviral proteins during infection results from an evasion mechanism exhibited by CHIKV that is dependent on widespread shutoff of cellular protein synthesis and a targeted block to late synthesis of antiviral mRNA transcripts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Vírus Chikungunya/patogenicidade , Regulação da Expressão Gênica , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Biossíntese de Proteínas/imunologia , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Células Cultivadas , Vírus Chikungunya/imunologia , Cricetinae , Fibroblastos/imunologia , Fibroblastos/virologia , Humanos , Interferon beta , Proteínas/genética , Proteínas/metabolismo
17.
Proteomics ; 11(5): 854-64, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21280226

RESUMO

The essential roles of the endovacuolar system in health and disease call for the development of new tools allowing a better understanding of the complex molecular machinery involved in endocytic processes. We took advantage of the floating properties of small latex beads (sLB) on a discontinuous sucrose gradient to isolate highly purified endosomes following internalization of small latex beads in J774 macrophages and bone marrow-derived dendritic cells (DC). We particularly focused on the isolation of macrophages early endosomes and late endosomes/lysosomes (LE/LYS) as well as the isolation of LE/LYS from immature and lipopolysaccharide-activated (mature) DC. We subsequently performed a comparative analysis of their respective protein contents by MS. As expected, proteins already known to localize to the early endosomes were enriched in the earliest fraction of J774 endosomes, while proteins known to accumulate later in the process, such as hydrolases, were significantly enriched in the LE/LYS preparations. We next compared the LE/LYS protein contents of immature DC and mature DC, which are known to undergo massive reorganization leading to potent immune activation. The differences between the protein contents of endocytic organelles from macrophages and DC were underlined by focusing on previously poorly characterized biochemical pathways, which could have an unexpected but important role in the endosomal functions of these highly relevant immune cell types.


Assuntos
Células Dendríticas/citologia , Endossomos/metabolismo , Macrófagos/citologia , Proteínas/metabolismo , Proteoma/metabolismo , Animais , Medula Óssea/imunologia , Medula Óssea/metabolismo , Fracionamento Celular/métodos , Linhagem Celular , Centrifugação com Gradiente de Concentração , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endocitose/efeitos dos fármacos , Endocitose/imunologia , Endossomos/química , Endossomos/imunologia , Lipopolissacarídeos/farmacologia , Lisossomos/química , Lisossomos/imunologia , Lisossomos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Microesferas , Fagossomos/imunologia , Fagossomos/metabolismo , Proteínas/análise , Proteínas/classificação , Proteínas/imunologia , Proteoma/análise , Proteoma/classificação , Proteoma/imunologia , Sacarose/química
18.
Curr Opin Cell Biol ; 15(4): 468-73, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12892788

RESUMO

The study of the cell biology of antigen processing and presentation has greatly contributed to our understanding of the immune response. The work of many immunologically inclined cell biologists has also permitted us to gain new insights on cellular mechanisms shared by many cell types. Dendritic cells are master regulators of the immune system and consequently have received a lot of attention in recent years. With the aim of controlling antigen processing and presentation, the solutions used by dendritic cells to respond to environmental changes are numerous and surprising. In the presence of pathogens, dendritic cells regulate strongly their endocytic pathway by interfering with uptake, proteolysis, membrane dynamics and transport in and out of the lysosome to become the most potent antigen-presenting cells known.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos/metabolismo , Células Dendríticas/imunologia , Endocitose/imunologia , Animais , Endossomos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Lisossomos/imunologia , Lisossomos/metabolismo , Transporte Proteico/imunologia
19.
Proc Natl Acad Sci U S A ; 105(9): 3491-6, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18305173

RESUMO

In response to Toll-like receptor ligands, dendritic cells (DCs) dramatically enhance their antigen presentation capacity by stabilizing at the cell-surface MHC II molecules. We demonstrate here that, in human monocyte-derived DCs, the RING-CH ubiquitin E3 ligase, membrane-associated RING-CH I (MARCH I), promotes the ubiquitination of the HLA-DR beta-chain. Thus, in nonactivated DCs, MARCH I induces the surface internalization of mature HLA-DR complexes, therefore reducing their stability and levels. We further demonstrate that the maturation-dependent down-regulation of MARCH I is a key event in MHC class II up-regulation at the surface of LPS-activated DCs. MARCH I is, therefore, a major regulator of HLA-DR traffic, and its loss contributes to the acquisition of the potent immunostimulatory properties of mature human DCs.


Assuntos
Células Dendríticas/imunologia , Regulação para Baixo/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Ubiquitina-Proteína Ligases/genética , Antígenos de Superfície , Transporte Biológico , Células Cultivadas , Endocitose , Antígenos HLA-DR/metabolismo , Humanos , Lipopolissacarídeos/farmacologia
20.
R Soc Open Sci ; 8(7): 202333, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34295519

RESUMO

We report here that RUFY4, a newly characterized member of the 'RUN and FYVE domain-containing' family of proteins previously associated with autophagy enhancement, is highly expressed in alveolar macrophages (AM). We show that RUFY4 interacts with mitochondria upon stimulation by microbial-associated molecular patterns of AM and dendritic cells. RUFY4 interaction with mitochondria and other organelles is dependent on a previously uncharacterized OmpH domain located immediately upstream of its C-terminal FYVE domain. Further, we demonstrate that rufy4 messenger RNA can be translated from an alternative translation initiation codon, giving rise to a N-terminally truncated form of the molecule lacking most of its RUN domain and with enhanced potential for its interaction with mitochondria. Our observations point towards a role of RUFY4 in selective mitochondria clearance in activated phagocytes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA