Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
PLoS Genet ; 18(4): e1009799, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35377871

RESUMO

Centrioles are submicron-scale, barrel-shaped organelles typically found in pairs, and play important roles in ciliogenesis and bipolar spindle assembly. In general, successful execution of centriole-dependent processes is highly reliant on the ability of the cell to stringently control centriole number. This in turn is mainly achieved through the precise duplication of centrioles during each S phase. Aberrations in centriole duplication disrupt spindle assembly and cilia-based signaling and have been linked to cancer, primary microcephaly and a variety of growth disorders. Studies aimed at understanding how centriole duplication is controlled have mainly focused on the post-translational regulation of two key components of this pathway: the master regulatory kinase ZYG-1/Plk4 and the scaffold component SAS-6. In contrast, how transcriptional control mechanisms might contribute to this process have not been well explored. Here we show that the chromatin remodeling protein CHD-1 contributes to the regulation of centriole duplication in the C. elegans embryo. Specifically, we find that loss of CHD-1 or inactivation of its ATPase activity can restore embryonic viability and centriole duplication to a strain expressing insufficient ZYG-1 activity. Interestingly, loss of CHD-1 is associated with increases in the levels of two ZYG-1-binding partners: SPD-2, the centriole receptor for ZYG-1 and SAS-6. Finally, we explore transcriptional regulatory networks governing centriole duplication and find that CHD-1 and a second transcription factor, EFL-1/DPL-1 cooperate to down regulate expression of CDK-2, which in turn promotes SAS-6 protein levels. Disruption of this regulatory network results in the overexpression of SAS-6 and the production of extra centrioles.


Assuntos
Proteínas de Caenorhabditis elegans , Centríolos , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Centríolos/genética , Centríolos/metabolismo , Montagem e Desmontagem da Cromatina/genética , Proteínas Quinases/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
2.
Mol Cell ; 52(4): 495-505, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24095279

RESUMO

The mTORC1 kinase is a master growth regulator that senses numerous environmental cues, including amino acids. The Rag GTPases interact with mTORC1 and signal amino acid sufficiency by promoting the translocation of mTORC1 to the lysosomal surface, its site of activation. The Rags are unusual GTPases in that they function as obligate heterodimers, which consist of RagA or B bound to RagC or D. While the loading of RagA/B with GTP initiates amino acid signaling to mTORC1, the role of RagC/D is unknown. Here, we show that RagC/D is a key regulator of the interaction of mTORC1 with the Rag heterodimer and that, unexpectedly, RagC/D must be GDP bound for the interaction to occur. We identify FLCN and its binding partners, FNIP1/2, as Rag-interacting proteins with GAP activity for RagC/D, but not RagA/B. Thus, we reveal a role for RagC/D in mTORC1 activation and a molecular function for the FLCN tumor suppressor.


Assuntos
Aminoácidos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Proteínas de Transporte/metabolismo , Proteínas Ativadoras de GTPase/fisiologia , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Ligação Proteica , Transporte Proteico , Transdução de Sinais
3.
J Biol Chem ; 294(28): 10746-10757, 2019 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-31076509

RESUMO

In eukaryotes, ribosome assembly is a rate-limiting step in ribosomal biogenesis that takes place in a distinctive subnuclear organelle, the nucleolus. How ribosomes get assembled at the nucleolar site by forming initial preribosomal complexes remains poorly characterized. In this study, using several human and murine cell lines, we developed a method for isolation of native mammalian preribosomal complexes by lysing cell nuclei through mild sonication. A sucrose gradient fractionation of the nuclear lysate resolved several ribonucleoprotein (RNP) complexes containing rRNAs and ribosomal proteins. Characterization of the RNP complexes with MS-based protein identification and Northern blotting-based rRNA detection approaches identified two types of preribosomes we named here as intermediate preribosomes (IPRibs) and composed preribosome (CPRib). IPRib complexes comprised large preribosomes (105S to 125S in size) containing the rRNA modification factors and premature rRNAs. We further observed that a distinctive CPRib complex consists of an 85S preribosome assembled with mature rRNAs and a ribosomal biogenesis factor, Ly1 antibody-reactive (LYAR), that does not associate with premature rRNAs and rRNA modification factors. rRNA-labeling experiments uncovered that IPRib assembly precedes CPRib complex formation. We also found that formation of the preribosomal complexes is nutrient-dependent because the abundances of IPRib and CPRib decreased substantially when cells were either deprived of amino acids or exposed to an mTOR kinase inhibitor. These findings indicate that preribosomes form via dynamic and nutrient-dependent processing events and progress from an intermediate to a composed state during ribosome maturation.


Assuntos
Precursores de RNA/metabolismo , Ribossomos/metabolismo , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Acetiltransferases N-Terminal/metabolismo , Processamento Pós-Transcricional do RNA , RNA Ribossômico/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Proteínas Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo
4.
Nat Immunol ; 9(12): 1407-14, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18931679

RESUMO

Toll-like receptors (TLRs) activate the innate immune system in response to pathogens. Here we show that TLR9 proteolytic cleavage is a prerequisite for TLR9 signaling. Inhibition of lysosomal proteolysis rendered TLR9 inactive. The carboxy-terminal fragment of TLR9 thus generated included a portion of the TLR9 ectodomain, as well as the transmembrane and cytoplasmic domains. This cleavage fragment bound to the TLR9 ligand CpG DNA and, when expressed in Tlr9(-/-) dendritic cells, restored CpG DNA-induced cytokine production. Although cathepsin L generated the requisite TLR9 cleavage products in a cell-free in vitro system, several proteases influenced TLR9 cleavage in intact cells. Lysosomal proteolysis thus contributes to innate immunity by facilitating specific cleavage of TLR9.


Assuntos
Ativação Enzimática/imunologia , Imunidade Inata/fisiologia , Lisossomos/metabolismo , Transdução de Sinais/imunologia , Receptor Toll-Like 9/metabolismo , Animais , Western Blotting , Catepsinas/imunologia , Catepsinas/metabolismo , Linhagem Celular , Ilhas de CpG , Eletroforese em Gel de Poliacrilamida , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunoprecipitação , Lisossomos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico/imunologia , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia
5.
Immunity ; 34(4): 505-13, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21497117

RESUMO

Toll-like receptor (TLR) signaling plays a critical role in innate and adaptive immune responses and must be tightly controlled. TLR4 uses LPS binding protein, MD-2, and CD14 as accessories to respond to LPS. We therefore investigated the presence of an analagous soluble cofactor that might assist in the recruitment of CpG oligonucleotides (CpG-ODNs) to TLR9. We report the identification of granulin as an essential secreted cofactor that potentiates TLR9-driven responses to CpG-ODNs. Granulin, an unusual cysteine-rich protein, bound to CpG-ODNs and interacted with TLR9. Macrophages from granulin-deficient mice showed not only impaired delivery of CpG-ODNs to endolysosomal compartments, but also decreased interaction of TLR9 with CpG-ODNs. As a consequence, granulin-deficient macrophages showed reduced responses to stimulation with CpG-ODNs, a trait corrected by provision of exogenous granulin. Thus, we propose that granulin contributes to innate immunity as a critical soluble cofactor for TLR9 signaling.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Transdução de Sinais , Receptor Toll-Like 9/imunologia , Animais , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodesoxirribonucleotídeos/imunologia , Progranulinas , Ligação Proteica , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo
6.
Proc Natl Acad Sci U S A ; 113(6): E705-14, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26811477

RESUMO

Posttranslational modifications are central to the spatial and temporal regulation of protein function. Among others, phosphorylation and ubiquitylation are known to regulate proximal T-cell receptor (TCR) signaling. Here we used a systematic and unbiased approach to uncover deubiquitylating enzymes (DUBs) that participate during TCR signaling in primary mouse T lymphocytes. Using a C-terminally modified vinyl methyl ester variant of ubiquitin (HA-Ub-VME), we captured DUBs that are differentially recruited to the cytosol on TCR activation. We identified ubiquitin-specific peptidase (Usp) 12 and Usp46, which had not been previously described in this pathway. Stimulation with anti-CD3 resulted in phosphorylation and time-dependent translocation of Usp12 from the nucleus to the cytosol. Usp12(-/-) Jurkat cells displayed defective NFκB, NFAT, and MAPK activities owing to attenuated surface expression of TCR, which were rescued on reconstitution of wild type Usp12. Proximity-based labeling with BirA-Usp12 revealed several TCR adaptor proteins acting as interactors in stimulated cells, of which LAT and Trat1 displayed reduced expression in Usp12(-/-) cells. We demonstrate that Usp12 deubiquitylates and prevents lysosomal degradation of LAT and Trat1 to maintain the proximal TCR complex for the duration of signaling. Our approach benefits from the use of activity-based probes in primary cells without any previous genome modification, and underscores the importance of ubiquitin-mediated regulation to refine signaling cascades.


Assuntos
Membrana Celular/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Ubiquitina Tiolesterase/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Núcleo Celular/metabolismo , Separação Celular , Citosol/metabolismo , Endopeptidases/metabolismo , Ácidos Graxos Insaturados/farmacologia , Humanos , Células Jurkat , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Transporte Proteico , Reprodutibilidade dos Testes , Especificidade por Substrato/efeitos dos fármacos , Linfócitos T/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterase/deficiência , Proteases Específicas de Ubiquitina/metabolismo
7.
J Immunol ; 192(12): 6028-36, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24821971

RESUMO

IL-12p40 partners with the p35 and p19 polypeptides to generate the heterodimeric cytokines IL-12 and IL-23, respectively. These cytokines play critical and distinct roles in host defense. The assembly of these heterodimers is thought to take place within the cell, resulting in the secretion of fully functional cytokines. Although the p40 subunit alone can also be rapidly secreted in response to inflammatory signals, its biological significance remains unclear. In this article, we show that the secreted p40 monomer can generate de novo IL-12-like activities by combining extracellularly with p35 released from other cells. Surprisingly, an unbiased proteomic analysis reveals multiple such extracellular binding partners for p40 in the serum of mice after an endotoxin challenge. We biochemically validate the binding of one of these novel partners, the CD5 Ag-like glycoprotein, to the p40 monomer. Nevertheless, the assembled p40-CD5L heterodimer does not recapitulate the biological activity of IL-12. These findings underscore the plasticity of secreted free p40 monomer, suggesting that p40 functions as an adaptor that is able to generate multiple de novo composites in combination with other locally available polypeptide partners after secretion.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Dimerização , Interleucina-12/imunologia , Receptores Imunológicos/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Antígenos CD5/genética , Antígenos CD5/imunologia , Interleucina-12/genética , Camundongos , Camundongos Knockout , Proteômica , Receptores Imunológicos/genética , Receptores Depuradores
8.
Proc Natl Acad Sci U S A ; 110(4): 1428-33, 2013 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-23297227

RESUMO

A monoclonal antibody against the C-type lectin DEC205 (αDEC205) is an effective vehicle for delivery of antigens to dendritic cells through creation of covalent αDEC205-antigen adducts. These adducts can induce antigen-specific T-cell immune responses or tolerance. We exploit the transpeptidase activity of sortase to install modified peptides and protein-sized antigens onto the heavy chain of αDEC205, including linkers that contain nonnatural amino acids. We demonstrate stoichiometric site-specific labeling on a scale not easily achievable by genetic fusions (49 distinct fusions in this report). We conjugated a biotinylated version of a class I MHC-restricted epitope to unlabeled αDEC205 and monitored epitope generation upon binding of the adduct to dendritic cells. Our results show transfer of αDEC205 heavy chain to the cytoplasm, followed by proteasomal degradation. Introduction of a labile dipeptide linker at the N terminus of a T-cell epitope improves proteasome-dependent class I MHC-restricted peptide cross-presentation when delivered by αDEC205 in vitro and in vivo. We also conjugated αDEC205 with a linker-optimized peptide library of known CD8 T-cell epitopes from the mouse γ-herpes virus 68. Animals immunized with such conjugates displayed a 10-fold reduction in viral load.


Assuntos
Aminoaciltransferases/metabolismo , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos Virais/imunologia , Proteínas de Bactérias/metabolismo , Cisteína Endopeptidases/metabolismo , Epitopos de Linfócito T/imunologia , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Animais , Apresentação de Antígeno , Antígenos CD/química , Antígenos CD/genética , Antígenos Virais/química , Antígenos Virais/genética , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/química , Epitopos de Linfócito T/genética , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunização , Imunoconjugados/genética , Imunoconjugados/imunologia , Imunoconjugados/metabolismo , Lectinas Tipo C/química , Lectinas Tipo C/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Antígenos de Histocompatibilidade Menor , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Engenharia de Proteínas , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Rhadinovirus/genética , Rhadinovirus/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/prevenção & controle , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia
9.
PLoS Pathog ; 9(6): e1003446, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825946

RESUMO

Phagocytosis of the opportunistic fungal pathogen Candida albicans by cells of the innate immune system is vital to prevent infection. Dectin-1 is the major phagocytic receptor involved in anti-fungal immunity. We identify two new interacting proteins of Dectin-1 in macrophages, Bruton's Tyrosine Kinase (BTK) and Vav1. BTK and Vav1 are recruited to phagocytic cups containing C. albicans yeasts or hyphae but are absent from mature phagosomes. BTK and Vav1 localize to cuff regions surrounding the hyphae, while Dectin-1 lines the full length of the phagosome. BTK and Vav1 colocalize with the lipid PI(3,4,5)P3 and F-actin at the phagocytic cup, but not with diacylglycerol (DAG) which marks more mature phagosomal membranes. Using a selective BTK inhibitor, we show that BTK contributes to DAG synthesis at the phagocytic cup and the subsequent recruitment of PKCε. BTK- or Vav1-deficient peritoneal macrophages display a defect in both zymosan and C. albicans phagocytosis. Bone marrow-derived macrophages that lack BTK or Vav1 show reduced uptake of C. albicans, comparable to Dectin1-deficient cells. BTK- or Vav1-deficient mice are more susceptible to systemic C. albicans infection than wild type mice. This work identifies an important role for BTK and Vav1 in immune responses against C. albicans.


Assuntos
Candida albicans/imunologia , Candidíase/imunologia , Proteínas de Homeodomínio/imunologia , Lectinas Tipo C/imunologia , Macrófagos Peritoneais/imunologia , Neuropeptídeos/imunologia , Fagocitose/imunologia , Proteínas Tirosina Quinases/imunologia , Actinas/genética , Actinas/imunologia , Actinas/metabolismo , Tirosina Quinase da Agamaglobulinemia , Animais , Candida albicans/metabolismo , Candidíase/genética , Candidíase/metabolismo , Candidíase/patologia , Linhagem Celular , Diglicerídeos/genética , Diglicerídeos/imunologia , Diglicerídeos/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Knockout , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fagocitose/genética , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/imunologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo
10.
J Immunol ; 191(1): 208-16, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23740953

RESUMO

Multiple sclerosis (MS) is an autoimmune disease that affects the CNS. One approved treatment for relapsing forms of MS is YEAK, a random copolymer of the amino acids tyrosine, glutamic acid, alanine, and lysine. YFAK, a second-generation copolymer composed of tyrosine, phenylalanine, alanine, and lysine, is more successful in treating experimental autoimmune encephalomyelitis, a mouse model of MS. Although originally designed and optimized based on the autoantigen myelin basic protein (MBP) and the MBP-derived peptide MBP85-99 presented to the MS-associated class II MHC molecule HLA-DR2, YEAK and YFAK also stimulate cytokine and chemokine production in APCs that lack class II MHC products. How YEAK and YFAK copolymers interact with APCs remains enigmatic. We used biotinylated YFAK to affinity-purify YFAK-interacting proteins from RAW264.7 cells and tested APCs from mice deficient in several of the newly identified interactors for their capacity to secrete CCL22 in response to YEAK and YFAK. We propose that initial contact of YFAK with cells is mediated mainly by electrostatic interactions, and find that interaction of YFAK with host proteins is strongly dependent on ionic strength. Cells deficient in enzymes involved in sulfation of proteins and proteoglycans showed strongly reduced binding of biotinylated YFAK. Lastly, cells stimulated with YFAK in the presence of heparin, structurally similar to heparan sulfates, failed to produce CCL22. We conclude that charge-dependent interactions of copolymers that alleviate MS/experimental autoimmune encephalomyelitis are critical for their effects exerted on APCs and may well be the main initial mediators of these therapeutically active copolymers.


Assuntos
Aminoácidos/metabolismo , Células Apresentadoras de Antígenos/química , Células Apresentadoras de Antígenos/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/prevenção & controle , Heparitina Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Mapeamento de Interação de Proteínas/métodos , Aminoácidos/química , Aminoácidos/farmacologia , Animais , Células Apresentadoras de Antígenos/imunologia , Biopolímeros/química , Biopolímeros/metabolismo , Biopolímeros/farmacologia , Biotinilação , Linhagem Celular , Encefalomielite Autoimune Experimental/metabolismo , Retículo Endoplasmático/genética , Retículo Endoplasmático/imunologia , Retículo Endoplasmático/metabolismo , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Cultura Primária de Células , Distribuição Aleatória
11.
Traffic ; 13(6): 780-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22348280

RESUMO

Sortagging is a versatile method for site-specific modification of proteins as applied to a variety of in vitro reactions. Here, we explore possibilities of adapting the sortase method for use in living cells. For intracellular sortagging, we employ the Ca²âº-independent sortase A transpeptidase (SrtA) from Streptococcus pyogenes. Substrate proteins were equipped with the C-terminal sortase-recognition motif (LPXTG); we used proteins with an N-terminal (oligo)glycine as nucleophiles. We show that sortase-dependent protein ligation can be achieved in Saccharomyces cerevisiae and in mammalian HEK293T cells, both in the cytosol and in the lumen of the endoplasmic reticulum (ER). ER luminal sortagging enables secretion of the reaction products, among which circular polypeptides. Protein ligation of substrate and nucleophile occurs within 30 min of translation. The versatility of the method is shown by protein ligation of multiple substrates with green fluorescent protein-based nucleophiles in different intracellular compartments.


Assuntos
Aminoaciltransferases/fisiologia , Proteínas de Bactérias/fisiologia , Cisteína Endopeptidases/fisiologia , Streptococcus pyogenes/metabolismo , Aminoaciltransferases/metabolismo , Proteínas de Bactérias/metabolismo , Biologia Celular , Cisteína Endopeptidases/metabolismo , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Cinética , Espectrometria de Massas/métodos , Peptídeos/química , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Saccharomyces cerevisiae/metabolismo
12.
J Biol Chem ; 288(38): 27019-27030, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23928304

RESUMO

Nutrients are essential for living organisms because they fuel biological processes in cells. Cells monitor nutrient abundance and coordinate a ratio of anabolic and catabolic reactions. Mechanistic target of rapamycin (mTOR) signaling is the essential nutrient-sensing pathway that controls anabolic processes in cells. The central component of this pathway is mTOR, a highly conserved and essential protein kinase that exists in two distinct functional complexes. The nutrient-sensitive mTOR complex 1 (mTORC1) controls cell growth and cell size by phosphorylation of the regulators of protein synthesis S6K1 and 4EBP1, whereas its second complex, mTORC2, regulates cell proliferation by functioning as the regulatory kinase of Akt and other members of the AGC kinase family. The regulation of mTORC2 remains poorly characterized. Our study shows that the cellular ATP balance controls a basal kinase activity of mTORC2 that maintains the integrity of mTORC2 and phosphorylation of Akt on the turn motif Thr-450 site. We found that mTOR stabilizes SIN1 by phosphorylation of its hydrophobic and conserved Ser-260 site to maintain the integrity of mTORC2. The optimal kinase activity of mTORC2 requires a concentration of ATP above 1.2 mM and makes this kinase complex highly sensitive to ATP depletion. We found that not amino acid but glucose deprivation of cells or acute ATP depletion prevented the mTOR-dependent phosphorylation of SIN1 on Ser-260 and Akt on Thr-450. In a low glucose medium, the cells carrying a substitution of SIN1 with its phosphomimetic mutant show an increased rate of cell proliferation related to a higher abundance of mTORC2 and phosphorylation of Akt. Thus, the homeostatic ATP sensor mTOR controls the integrity of mTORC2 and phosphorylation of Akt on the turn motif site.


Assuntos
Trifosfato de Adenosina/metabolismo , Proliferação de Células , Complexos Multiproteicos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Trifosfato de Adenosina/genética , Motivos de Aminoácidos , Células HeLa , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Complexos Multiproteicos/genética , Mutação , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/genética
13.
Infect Immun ; 82(2): 706-19, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478085

RESUMO

The gamma interferon (IFN-γ) response, mediated by the STAT1 transcription factor, is crucial for host defense against the intracellular pathogen Toxoplasma gondii, but prior infection with Toxoplasma can inhibit this response. Recently, it was reported that the Toxoplasma type II NTE strain prevents the recruitment of chromatin remodeling complexes containing Brahma-related gene 1 (BRG-1) to promoters of IFN-γ-induced secondary response genes such as Ciita and major histocompatibility complex class II genes in murine macrophages, thereby inhibiting their expression. We report here that a type I strain of Toxoplasma inhibits the expression of primary IFN-γ response genes such as IRF1 through a distinct mechanism not dependent on the activity of histone deacetylases. Instead, infection with a type I, II, or III strain of Toxoplasma inhibits the dissociation of STAT1 from DNA, preventing its recycling and further rounds of STAT1-mediated transcriptional activation. This leads to increased IFN-γ-induced binding of STAT1 at the IRF1 promoter in host cells and increased global IFN-γ-induced association of STAT1 with chromatin. Toxoplasma type I infection also inhibits IFN-ß-induced interferon-stimulated gene factor 3-mediated gene expression, and this inhibition is also linked to increased association of STAT1 with chromatin. The secretion of proteins into the host cell by a type I strain of Toxoplasma without complete parasite invasion is not sufficient to block STAT1-mediated expression, suggesting that the effector protein responsible for this inhibition is not derived from the rhoptries.


Assuntos
DNA/metabolismo , Interações Hospedeiro-Patógeno , Interferon beta/antagonistas & inibidores , Interferon gama/antagonistas & inibidores , Fator de Transcrição STAT1/metabolismo , Toxoplasma/imunologia , Linhagem Celular , Humanos , Evasão da Resposta Imune , Interferon beta/imunologia , Interferon gama/imunologia , Ligação Proteica
14.
PLoS Pathog ; 8(6): e1002784, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761577

RESUMO

The obligate intracellular parasite Toxoplasma gondii secretes effector proteins into the host cell that manipulate the immune response allowing it to establish a chronic infection. Crosses between the types I, II and III strains, which are prevalent in North America and Europe, have identified several secreted effectors that determine strain differences in mouse virulence. The polymorphic rhoptry protein kinase ROP18 was recently shown to determine the difference in virulence between type I and III strains by phosphorylating and inactivating the interferon-γ (IFNγ)-induced immunity-related GTPases (IRGs) that promote killing by disrupting the parasitophorous vacuole membrane (PVM) in murine cells. The polymorphic pseudokinase ROP5 determines strain differences in virulence through an unknown mechanism. Here we report that ROP18 can only inhibit accumulation of the IRGs on the PVM of strains that also express virulent ROP5 alleles. In contrast, specific ROP5 alleles can reduce IRG coating even in the absence of ROP18 expression and can directly interact with one or more IRGs. We further show that the allelic combination of ROP18 and ROP5 also determines IRG evasion and virulence of strains belonging to other lineages besides types I, II and III. However, neither ROP18 nor ROP5 markedly affect survival in IFNγ-activated human cells, which lack the multitude of IRGs present in murine cells. These findings suggest that ROP18 and ROP5 have specifically evolved to block the IRGs and are unlikely to have effects in species that do not have the IRG system, such as humans.


Assuntos
Evasão da Resposta Imune/imunologia , Interferon gama/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Animais , Western Blotting , Cromatografia Líquida de Alta Pressão , Imunofluorescência , GTP Fosfo-Hidrolases , Ensaios de Triagem em Larga Escala , Humanos , Imunoprecipitação , Espectrometria de Massas , Camundongos , Proteínas de Protozoários , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Especificidade da Espécie , Toxoplasma/patogenicidade , Virulência/imunologia
15.
PLoS Biol ; 8(3): e1000605, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21468303

RESUMO

Ubiquitin-dependent processes control much of cellular physiology. We show that expression of a highly active, Epstein-Barr virus-derived deubiquitylating enzyme (EBV-DUB) blocks proteasomal degradation of cytosolic and ER-derived proteins by preemptive removal of ubiquitin from proteasome substrates, a treatment less toxic than the use of proteasome inhibitors. Recognition of misfolded proteins in the ER lumen, their dislocation to the cytosol, and degradation are usually tightly coupled but can be uncoupled by the EBV-DUB: a misfolded glycoprotein that originates in the ER accumulates in association with cytosolic chaperones as a deglycosylated intermediate. Our data underscore the necessity of a DUB activity for completion of the dislocation reaction and provide a new means of inhibition of proteasomal proteolysis with reduced cytotoxicity.


Assuntos
Herpesvirus Humano 4/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Proteínas Virais/metabolismo , Biocatálise , Linhagem Celular , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Processamento de Proteína Pós-Traducional , Transporte Proteico , Especificidade por Substrato
16.
Proc Natl Acad Sci U S A ; 108(8): 3169-74, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21297034

RESUMO

Recombinant protein therapeutics often suffer from short circulating half-life and poor stability, necessitating multiple injections and resulting in limited shelf-life. Conjugation to polyethylene glycol chains (PEG) extends the circulatory half-life of many proteins, but the methods for attachment often lack specificity, resulting in loss of biological activity. Using four-helix bundle cytokines as an example, we present a general platform that uses sortase-mediated transpeptidation to facilitate site-specific attachment of PEG to extend cytokine half-life with full retention of biological activity. Covalently joining the N and C termini of proteins to obtain circular polypeptides, again executed using sortase, increases thermal stability. We combined both PEGylation and circularization by exploiting two distinct sortase enzymes and the use of a molecular suture that allows both site-specific PEGylation and covalent closure. The method developed is general, uses a set of easily accessible reagents, and should be applicable to a wide variety of proteins, provided that their termini are not involved in receptor binding or function.


Assuntos
Aminoaciltransferases/química , Proteínas de Bactérias/química , Cisteína Endopeptidases/química , Polietilenoglicóis/química , Estabilidade Proteica , Aminoaciltransferases/administração & dosagem , Aminoaciltransferases/metabolismo , Animais , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/metabolismo , Catálise , Ciclização , Cisteína Endopeptidases/administração & dosagem , Cisteína Endopeptidases/metabolismo , Citocinas , Meia-Vida , Métodos , Camundongos , Streptococcus pyogenes
17.
Proc Natl Acad Sci U S A ; 108(22): 9238-43, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21562218

RESUMO

Type I strains of Helicobacter pylori (Hp) possess a pathogenicity island, cag, that encodes the effector protein cytotoxin-associated gene A (CagA) and a type four secretion system. After translocation into the host cell, CagA affects cell shape, increases cell motility, abrogates junctional activity, and promotes an epithelial to mesenchymal transition-like phenotype. Transgenic expression of CagA enhances gastrointestinal and intestinal carcinomas as well as myeloid and B-cell lymphomas in mice, but the mechanism of the induced cancer formation is not fully understood. Here, we show that CagA subverts the tumor suppressor function of apoptosis-stimulating protein of p53 (ASPP2). Delivery of CagA inside the host results in its association with ASPP2. After this interaction, ASPP2 recruits its natural target p53 and inhibits its apoptotic function. CagA leads to enhanced degradation of p53 and thereby, down-regulates its activity in an ASPP2-dependent manner. Finally, Hp-infected cells treated with the p53-activating drug Doxorubicin are more resistant to apoptosis than uninfected cells, an effect that requires ASPP2. The interaction between CagA and ASPP2 and the consequent degradation of p53 are examples of a bacterial protein that subverts the p53 tumor suppressor pathway in a manner similar to DNA tumor viruses. This finding may contribute to the understanding of the increased risk of gastric cancer in patients infected with Hp CagA+ strains.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Proteínas de Bactérias/metabolismo , Regulação da Expressão Gênica , Helicobacter pylori/metabolismo , Sequência de Aminoácidos , DNA/metabolismo , Doxorrubicina/farmacologia , Genes Supressores de Tumor , Humanos , Dados de Sequência Molecular , Fenótipo , Risco , Neoplasias Gástricas/metabolismo , Proteína Supressora de Tumor p53/metabolismo
18.
Proc Natl Acad Sci U S A ; 108(5): 1763-70, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21209336

RESUMO

The ubiquitin (Ub)-related modifier Urm1 functions as a sulfur carrier in tRNA thiolation by means of a mechanism that requires the formation of a thiocarboxylate at the C-terminal glycine residue of Urm1. However, whether Urm1 plays an additional role as a Ub-like protein modifier remains unclear. Here, we show that Urm1 is conjugated to lysine residues of target proteins and that oxidative stress enhances protein urmylation in both Saccharomyces cerevisiae and mammalian cells. Similar to ubiquitylation, urmylation involves a thioester intermediate and results in the formation of a covalent peptide bond between Urm1 and its substrates. In contrast to modification by canonical Ub-like modifiers, however, conjugation of Urm1 involves a C-terminal thiocarboxylate of the modifier. We have confirmed that the peroxiredoxin Ahp1 is such a substrate in S. cerevisiae and found that Urm1 targets a specific lysine residue of Ahp1 in vivo. In addition, we have identified several unique substrates in mammalian cells and show that Urm1 targets at least two pathways on oxidant treatment. First, Urm1 is appended to lysine residues of three components that function in its own pathway (i.e., MOCS3, ATPBD3, and CTU2). Second, Urm1 is conjugated to the nucleocytoplasmic shuttling factor cellular apoptosis susceptibility protein. Thus, Urm1 has a conserved dual role by integrating the functions of prokaryotic sulfur carriers with those of eukaryotic protein modifiers of the Ub family.


Assuntos
Lisina/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Ubiquitinas/fisiologia , Cromatografia Líquida , Diamida/metabolismo , Eletroforese em Gel de Poliacrilamida , Humanos , Estresse Oxidativo , Proteômica , Proteínas de Saccharomyces cerevisiae/metabolismo , Espectrometria de Massas em Tandem , Ubiquitinação , Ubiquitinas/metabolismo
19.
Nat Cell Biol ; 26(4): 593-603, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38553595

RESUMO

Loss of protein function is a driving force of ageing. We have identified peptidyl-prolyl isomerase A (PPIA or cyclophilin A) as a dominant chaperone in haematopoietic stem and progenitor cells. Depletion of PPIA accelerates stem cell ageing. We found that proteins with intrinsically disordered regions (IDRs) are frequent PPIA substrates. IDRs facilitate interactions with other proteins or nucleic acids and can trigger liquid-liquid phase separation. Over 20% of PPIA substrates are involved in the formation of supramolecular membrane-less organelles. PPIA affects regulators of stress granules (PABPC1), P-bodies (DDX6) and nucleoli (NPM1) to promote phase separation and increase cellular stress resistance. Haematopoietic stem cell ageing is associated with a post-transcriptional decrease in PPIA expression and reduced translation of IDR-rich proteins. Here we link the chaperone PPIA to the synthesis of intrinsically disordered proteins, which indicates that impaired protein interaction networks and macromolecular condensation may be potential determinants of haematopoietic stem cell ageing.


Assuntos
Proteínas Intrinsicamente Desordenadas , Proteínas Intrinsicamente Desordenadas/química , Ciclofilina A/genética , Ciclofilina A/metabolismo , Proteínas de Ligação a RNA , Células-Tronco Hematopoéticas/metabolismo
20.
Chembiochem ; 14(3): 343-52, 2013 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-23335262

RESUMO

Protein ubiquitylation controls many cellular pathways, and timely removal of ubiquitin by deubiquitylating enzymes (DUBs) is essential to govern these different functions. To map endogenous expression of individual DUBs as well as that of any interacting proteins, we developed a catch-and-release ubiquitin probe. Ubiquitin was equipped with an activity-based warhead and a cleavable linker attached to a biotin affinity-handle through tandem site-specific modification, in which we combined intein chemistry with sortase-mediated ligation. The resulting probe is cell-impermeable and was therefore delivered to the cytosol of perfringolysin O (PFO)-permeabilized cells. This allowed us to retrieve and identify 34 DUBs and their interacting partners. We also noted the expression, in host cells infected with Chlamydia trachomatis, of two additional DUBs. Furthermore, we retrieved and identified chlamydial DUB1 (ChlaDUB1) and DUB2 (ChlaDUB2), demonstrating by experiment that ChlaDUB2, the presence and activity of which had not been detected in infected cells, is in fact expressed during the course of infection.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/metabolismo , Endopeptidases/metabolismo , Sondas Moleculares/metabolismo , Ubiquitina/química , Aminoaciltransferases/metabolismo , Compostos Azo/química , Proteínas de Bactérias/análise , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Biotina/química , Biotina/metabolismo , Infecções por Chlamydia/patologia , Chlamydia trachomatis/enzimologia , Cisteína Endopeptidases/metabolismo , Endopeptidases/análise , Endopeptidases/genética , Células HEK293 , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Humanos , Hidrazonas/química , Ácidos Levulínicos/química , Sondas Moleculares/química , Ubiquitina/metabolismo , Proteases Específicas de Ubiquitina , Ubiquitinação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA