Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Cell Biol ; 26(4): 593-603, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38553595

RESUMEN

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.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Proteínas Intrínsecamente Desordenadas/química , Ciclofilina A/genética , Ciclofilina A/metabolismo , Proteínas de Unión al ARN , Células Madre Hematopoyéticas/metabolismo
2.
PLoS Genet ; 18(4): e1009799, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35377871

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans , Centriolos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/genética , Centriolos/genética , Centriolos/metabolismo , Ensamble y Desensamble de Cromatina/genética , Proteínas Quinasas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Nat Commun ; 11(1): 5189, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060596

RESUMEN

Among the various host cellular processes that are hijacked by flaviviruses, few mechanisms have been described with regard to viral egress. Here we investigate how flaviviruses exploit Src family kinases (SFKs) for exit from infected cells. We identify Lyn as a critical component for secretion of Dengue and Zika infectious particles and their corresponding virus like particles (VLPs). Pharmacological inhibition or genetic depletion of the SFKs, Lyn in particular, block virus secretion. Lyn-/- cells are impaired in virus release and are rescued when reconstituted with wild-type Lyn, but not a kinase- or palmitoylation-deficient Lyn mutant. We establish that virus particles are secreted in two distinct populations - one as free virions and the other enclosed within membranes. Lyn is critical for the latter, which consists of proteolytically processed, infectious virus progenies within autophagosome-derived vesicles. This process depends on Ulk1, Rab GTPases and SNARE complexes implicated in secretory but not degradative autophagy and occur with significantly faster kinetics than the conventional secretory pathway. Our study reveals a previously undiscovered Lyn-dependent exit route of flaviviruses in LC3+ secretory organelles that enables them to evade circulating antibodies and might affect tissue tropism.


Asunto(s)
Autofagosomas/metabolismo , Autofagosomas/virología , Flavivirus/metabolismo , Familia-src Quinasas/metabolismo , Animales , Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular , Chlorocebus aethiops , Dengue , Virus del Dengue/metabolismo , Interacciones Microbiota-Huesped/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas SNARE/metabolismo , Vías Secretoras , Células Vero , Virión/metabolismo , Liberación del Virus , Virus Zika/metabolismo , Infección por el Virus Zika , Proteínas de Unión al GTP rab/metabolismo , Familia-src Quinasas/genética
4.
J Biol Chem ; 294(28): 10746-10757, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31076509

RESUMEN

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.


Asunto(s)
Precursores del ARN/metabolismo , Ribosomas/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Acetiltransferasas N-Terminal/metabolismo , Procesamiento Postranscripcional del ARN , ARN Ribosómico/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Proteínas Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
5.
Proc Natl Acad Sci U S A ; 113(6): E705-14, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26811477

RESUMEN

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.


Asunto(s)
Membrana Celular/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Ubiquitina Tiolesterasa/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Núcleo Celular/metabolismo , Separación Celular , Citosol/metabolismo , Endopeptidasas/metabolismo , Ácidos Grasos Insaturados/farmacología , Humanos , Células Jurkat , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Biológicos , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Transporte de Proteínas , Reproducibilidad de los Resultados , Especificidad por Sustrato/efectos de los fármacos , Linfocitos T/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/deficiencia , Proteasas Ubiquitina-Específicas/metabolismo
6.
PLoS One ; 9(10): e109883, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25275512

RESUMEN

Aerolysin is a secreted bacterial toxin that perforates the plasma membrane of a target cell with lethal consequences. Previously explored native and epitope-tagged forms of the toxin do not allow site-specific modification of the mature toxin with a probe of choice. We explore sortase-mediated transpeptidation reactions (sortagging) to install fluorophores and biotin at three distinct sites in aerolysin, without impairing binding of the toxin to the cell membrane and with minimal impact on toxicity. Using a version of aerolysin labeled with different fluorophores at two distinct sites we followed the fate of the C-terminal peptide independently from the N-terminal part of the toxin, and show its loss in the course of intoxication. Making use of the biotinylated version of aerolysin, we identify mesothelin, urokinase plasminogen activator surface receptor (uPAR, CD87), glypican-1, and CD59 glycoprotein as aerolysin receptors, all predicted or known to be modified with a glycosylphosphatidylinositol anchor. The sortase-mediated reactions reported here can be readily extended to other pore forming proteins.


Asunto(s)
Aeromonas hydrophila/metabolismo , Toxinas Bacterianas/metabolismo , Antígenos CD59/metabolismo , Glipicanos/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Aeromonas hydrophila/química , Secuencia de Aminoácidos , Animales , Toxinas Bacterianas/análisis , Biotina/química , Antígenos CD59/análisis , Línea Celular , Colorantes Fluorescentes/química , Glipicanos/análisis , Humanos , Datos de Secuencia Molecular , Imagen Óptica , Proteínas Citotóxicas Formadoras de Poros/análisis , Receptores del Activador de Plasminógeno Tipo Uroquinasa/análisis
7.
Cell Rep ; 9(1): 1-8, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25263562

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) kinase is a major regulator of cell growth that responds to numerous environmental cues. A key input is amino acids, which act through the heterodimeric Rag GTPases (RagA or RagB bound to RagC or RagD) in order to promote the translocation of mTORC1 to the lysosomal surface, its site of activation. GATOR2 is a complex of unknown function that positively regulates mTORC1 signaling by acting upstream of or in parallel to GATOR1, which is a GTPase-activating protein (GAP) for RagA or RagB and an inhibitor of the amino-acid-sensing pathway. Here, we find that the Sestrins, a family of poorly understood growth regulators (Sestrin1-Sestrin3), interact with GATOR2 in an amino-acid-sensitive fashion. Sestrin2-mediated inhibition of mTORC1 signaling requires GATOR1 and the Rag GTPases, and the Sestrins regulate the localization of mTORC1 in response to amino acids. Thus, we identify the Sestrins as GATOR2-interacting proteins that regulate the amino-acid-sensing branch of the mTORC1 pathway.


Asunto(s)
Aminoácidos/metabolismo , Proteínas de Choque Térmico/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células HEK293 , Proteínas de Choque Térmico/genética , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
8.
J Immunol ; 192(12): 6028-36, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24821971

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Dimerización , Interleucina-12/inmunología , Receptores Inmunológicos/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Antígenos CD5/genética , Antígenos CD5/inmunología , Interleucina-12/genética , Ratones , Ratones Noqueados , Proteómica , Receptores Inmunológicos/genética , Receptores Depuradores
9.
Infect Immun ; 82(2): 706-19, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478085

RESUMEN

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.


Asunto(s)
ADN/metabolismo , Interacciones Huésped-Patógeno , Interferón beta/antagonistas & inhibidores , Interferón gamma/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Toxoplasma/inmunología , Línea Celular , Humanos , Evasión Inmune , Interferón beta/inmunología , Interferón gamma/inmunología , Unión Proteica
10.
Cell Rep ; 5(5): 1302-15, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24290752

RESUMEN

Autophagy dysfunction has been implicated in misfolded protein accumulation and cellular toxicity in several diseases. Whether alterations in autophagy also contribute to the pathology of lipid-storage disorders is not clear. Here, we show defective autophagy in Niemann-Pick type C1 (NPC1) disease associated with cholesterol accumulation, where the maturation of autophagosomes is impaired because of defective amphisome formation caused by failure in SNARE machinery, whereas the lysosomal proteolytic function remains unaffected. Expression of functional NPC1 protein rescues this defect. Inhibition of autophagy also causes cholesterol accumulation. Compromised autophagy was seen in disease-affected organs of Npc1 mutant mice. Of potential therapeutic relevance is that HP-ß-cyclodextrin, which is used for cholesterol-depletion treatment, impedes autophagy, whereas stimulating autophagy restores its function independent of amphisome formation. Our data suggest that a low dose of HP-ß-cyclodextrin that does not perturb autophagy, coupled with an autophagy inducer, may provide a rational treatment strategy for NPC1 disease.


Asunto(s)
Autofagia , Glicoproteínas de Membrana/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Animales , Células Cultivadas , Colesterol/deficiencia , Colesterol/metabolismo , Células HEK293 , Humanos , Lisosomas/metabolismo , Glicoproteínas de Membrana/genética , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Ratas , Proteínas SNARE/metabolismo , beta-Ciclodextrinas/farmacología
11.
Mol Cell ; 52(4): 495-505, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24095279

RESUMEN

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.


Asunto(s)
Aminoácidos/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/fisiología , Proteínas Portadoras/metabolismo , Proteínas Activadoras de GTPasa/fisiología , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Lisosomas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Unión Proteica , Transporte de Proteínas , Transducción de Señal
12.
J Biol Chem ; 288(38): 27019-27030, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23928304

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proliferación Celular , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfato/genética , Secuencias de Aminoácidos , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/genética , Mutación , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética
13.
PLoS Pathog ; 9(6): e1003446, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825946

RESUMEN

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.


Asunto(s)
Candida albicans/inmunología , Candidiasis/inmunología , Proteínas de Homeodominio/inmunología , Lectinas Tipo C/inmunología , Macrófagos Peritoneales/inmunología , Neuropéptidos/inmunología , Fagocitosis/inmunología , Proteínas Tirosina Quinasas/inmunología , Actinas/genética , Actinas/inmunología , Actinas/metabolismo , Agammaglobulinemia Tirosina Quinasa , Animales , Candida albicans/metabolismo , Candidiasis/genética , Candidiasis/metabolismo , Candidiasis/patología , Línea Celular , Diglicéridos/genética , Diglicéridos/inmunología , Diglicéridos/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Ratones , Ratones Noqueados , Neuropéptidos/genética , Neuropéptidos/metabolismo , Fagocitosis/genética , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/inmunología , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo
14.
J Immunol ; 191(1): 208-16, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23740953

RESUMEN

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.


Asunto(s)
Aminoácidos/metabolismo , Células Presentadoras de Antígenos/química , Células Presentadoras de Antígenos/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Heparitina Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Mapeo de Interacción de Proteínas/métodos , Aminoácidos/química , Aminoácidos/farmacología , Animales , Células Presentadoras de Antígenos/inmunología , Biopolímeros/química , Biopolímeros/metabolismo , Biopolímeros/farmacología , Biotinilación , Línea Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligopéptidos/química , Oligopéptidos/metabolismo , Oligopéptidos/farmacología , Cultivo Primario de Células , Distribución Aleatoria
15.
ACS Synth Biol ; 2(9): 490-6, 2013 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-23713956

RESUMEN

M13 bacteriophage has been used as a scaffold to organize materials for various applications. Building more complex multiphage devices requires precise control of interactions between the M13 capsid proteins. Toward this end, we engineered a loop structure onto the pIII capsid protein of M13 bacteriophage to enable sortase-mediated labeling reactions for C-terminal display. Combining this with N-terminal sortase-mediated labeling, we thus created a phage scaffold that can be labeled orthogonally on three capsid proteins: the body and both ends. We show that covalent attachment of different DNA oligonucleotides at the ends of the new phage structure enables formation of multiphage particles oriented in a specific order. These have potential as nanoscale scaffolds for multi-material devices.


Asunto(s)
Bacteriófago M13/genética , Bacteriófago M13/metabolismo , Proteínas de la Cápside/química , ADN Viral/metabolismo , Secuencias de Aminoácidos , Bacteriófago M13/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Hibridación de Ácido Nucleico/métodos , Imagen Óptica , Ingeniería de Proteínas
16.
Chembiochem ; 14(3): 343-52, 2013 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-23335262

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Infecciones por Chlamydia/metabolismo , Endopeptidasas/metabolismo , Sondas Moleculares/metabolismo , Ubiquitina/química , Aminoaciltransferasas/metabolismo , Compuestos Azo/química , Proteínas Bacterianas/análisis , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/metabolismo , Biotina/química , Biotina/metabolismo , Infecciones por Chlamydia/patología , Chlamydia trachomatis/enzimología , Cisteína Endopeptidasas/metabolismo , Endopeptidasas/análisis , Endopeptidasas/genética , Células HEK293 , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Humanos , Hidrazonas/química , Ácidos Levulínicos/química , Sondas Moleculares/química , Ubiquitina/metabolismo , Proteasas Ubiquitina-Específicas , Ubiquitinación
17.
Proc Natl Acad Sci U S A ; 110(4): 1428-33, 2013 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-23297227

RESUMEN

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.


Asunto(s)
Aminoaciltransferasas/metabolismo , Antígenos CD/inmunología , Antígenos CD/metabolismo , Antígenos Virales/inmunología , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Epítopos de Linfocito T/inmunología , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Antígenos CD/química , Antígenos CD/genética , Antígenos Virales/química , Antígenos Virales/genética , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/química , Epítopos de Linfocito T/genética , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunización , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Inmunoconjugados/metabolismo , Lectinas Tipo C/química , Lectinas Tipo C/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Complejo de la Endopetidasa Proteasomal/metabolismo , Ingeniería de Proteínas , Receptores de Superficie Celular/química , Receptores de Superficie Celular/genética , Rhadinovirus/genética , Rhadinovirus/inmunología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/prevención & control , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
18.
PLoS One ; 7(10): e46709, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071619

RESUMEN

Inclusion body myositis (IBM) belongs to a group of muscle diseases known as the inflammatory myopathies. The presence of antibody-secreting plasma cells in IBM muscle implicates the humoral immune response in this disease. However, whether the humoral immune response actively contributes to IBM pathology has not been established. We sought to investigate whether the humoral immune response in IBM both in the periphery and at the site of tissue damage was directed towards self-antigens. Peripheral autoantibodies present in IBM serum but not control serum recognized self-antigens in both muscle tissue and human-derived cell lines. To study the humoral immune response at the site of tissue damage in IBM patients, we isolated single plasma cells directly from IBM-derived muscle tissue sections and from these cells, reconstructed a series of recombinant immunoglobulins (rIgG). These rIgG, each representing a single muscle-associated plasma cell, were examined for reactivity to self-antigens. Both, flow cytometry and immunoblotting revealed that these rIgG recognized antigens expressed by cell lines and in muscle tissue homogenates. Using a mass spectrometry-based approach, Desmin, a major intermediate filament protein, expressed abundantly in muscle tissue, was identified as the target of one IBM muscle-derived rIgG. Collectively, these data support the view that IBM includes a humoral immune response in both the periphery and at the site of tissue damage that is directed towards self-antigens.


Asunto(s)
Autoanticuerpos/sangre , Autoinmunidad , Inmunidad Humoral , Inmunoglobulina G/sangre , Miositis por Cuerpos de Inclusión/inmunología , Animales , Autoanticuerpos/química , Autoanticuerpos/aislamiento & purificación , Autoantígenos/inmunología , Autoantígenos/aislamiento & purificación , Estudios de Casos y Controles , Línea Celular , Desmina/inmunología , Desmina/aislamiento & purificación , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/aislamiento & purificación , Ratones , Proteínas Musculares/inmunología , Proteínas Musculares/aislamiento & purificación , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miositis por Cuerpos de Inclusión/sangre , Unión Proteica
19.
PLoS Pathog ; 8(6): e1002784, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761577

RESUMEN

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.


Asunto(s)
Evasión Inmune/inmunología , Interferón gamma/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Técnica del Anticuerpo Fluorescente , GTP Fosfohidrolasas , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunoprecipitación , Espectrometría de Masas , Ratones , Proteínas Protozoarias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Toxoplasma/patogenicidad , Virulencia/inmunología
20.
Bioconjug Chem ; 23(7): 1478-87, 2012 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-22759232

RESUMEN

We exploit bacterial sortases to attach a variety of moieties to the capsid proteins of M13 bacteriophage. We show that pIII, pIX, and pVIII can be functionalized with entities ranging from small molecules (e.g., fluorophores, biotin) to correctly folded proteins (e.g., GFP, antibodies, streptavidin) in a site-specific manner, and with yields that surpass those of any reported using phage display technology. A case in point is modification of pVIII. While a phage vector limits the size of the insert into pVIII to a few amino acids, a phagemid system limits the number of copies actually displayed at the surface of M13. Using sortase-based reactions, a 100-fold increase in the efficiency of display of GFP onto pVIII is achieved. Taking advantage of orthogonal sortases, we can simultaneously target two distinct capsid proteins in the same phage particle and maintain excellent specificity of labeling. As demonstrated in this work, this is a simple and effective method for creating a variety of structures, thus expanding the use of M13 for materials science applications and as a biological tool.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Bacteriófago M13/metabolismo , Proteínas de la Cápside/metabolismo , Técnicas de Visualización de Superficie Celular/métodos , Cisteína Endopeptidasas/metabolismo , Animales , Bacteriófago M13/química , Bacteriófago M13/genética , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Ratones , Ratones Endogámicos C57BL , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...