Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
RNA Biol ; 21(1): 1-17, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38551131

RESUMEN

RNA-binding proteins (RBPs) play crucial roles in the functions and homoeostasis of various tissues by regulating multiple events of RNA processing including RNA splicing, intracellular RNA transport, and mRNA translation. The Drosophila behavior and human splicing (DBHS) family proteins including PSF/SFPQ, NONO, and PSPC1 are ubiquitously expressed RBPs that contribute to the physiology of several tissues. In mammals, DBHS proteins have been reported to contribute to neurological diseases and play crucial roles in cancers, such as prostate, breast, and liver cancers, by regulating cancer-specific gene expression. Notably, in recent years, multiple small molecules targeting DBHS family proteins have been developed for application as cancer therapeutics. This review provides a recent overview of the functions of DBHS family in physiology and pathophysiology, and discusses the application of DBHS family proteins as promising diagnostic and therapeutic targets for cancers.


Asunto(s)
Drosophila , Neoplasias , Masculino , Animales , Humanos , Drosophila/genética , Drosophila/metabolismo , Proteínas de Unión al ARN/metabolismo , Empalme del ARN , ARN/metabolismo , Neoplasias/genética , Factor de Empalme Asociado a PTB/metabolismo , Mamíferos/genética
2.
Microb Pathog ; 185: 106391, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37839762

RESUMEN

Streptococcus pneumoniae colonizes the human nasopharynx asymptomatically, but it can also cause several diseases, including otitis media, pneumonia, bacteremia, and meningitis. The colonization of the nasopharynx by the bacteria is an essential step for the pneumococcus to invade other sites and cause diseases. Pneumococcal surface protein A (PspA) and Pneumococcal surface Protein C (PspC) are important virulence factors and have been described to play roles in adhesion and immune evasion. In this study, we immunized mice subcutaneously with the recombinant α-helical region of PspA and/or PspC combined with different adjuvants to assess protection against colonization with the serotype 6B strain BHN418. Though high serum levels of specific IgG were detected, none of the formulations led to reduction in the colonization of the nasopharynx. The negative result may be due to the poor induction of IgG2c, which has been previously correlated with protection against pneumococcal colonization in mice. Furthermore, BHN418 pspA and pspC single and double knockouts were evaluated in colonization experiments and no differences in bacterial load were observed. In competition assays with the wild-type strain, borderline to no reduction was observed in the loads of the knockouts. Our results contrast with data from the literature using other pneumococcal strains, showing that the role of PspA and PspC in colonization can vary depending on the background of the knockout strain studied. BHN418 has been selected for its capacity to colonize humans in experimental challenge studies and may have redundant factors that compensate for the lack of PspA and PspC during nasopharyngeal colonization of mice.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Animales , Ratones , Humanos , Infecciones Neumocócicas/microbiología , Proteína C/metabolismo , Serogrupo , Proteínas Bacterianas/metabolismo , Nasofaringe/microbiología , Proteínas de la Membrana/metabolismo , Vacunas Neumococicas , Anticuerpos Antibacterianos
3.
Int J Mol Sci ; 24(23)2023 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-38069409

RESUMEN

Poly (ADP-ribose) polymerase (PARP) inhibitors are effective against BRCA1/2-mutated cancers through synthetic lethality. Unfortunately, most cases ultimately develop acquired resistance. Therefore, enhancing PARP inhibitor sensitivity and preventing resistance in those cells are an unmet clinical need. Here, we investigated the ability of paraspeckle component 1 (PSPC1), as an additional synthetic lethal partner with BRCA1/2, to enhance olaparib sensitivity in preclinical models of BRCA1/2-mutated breast and ovarian cancers. In vitro, the combined olaparib and PSPC1 small interfering RNA (siRNA) exhibited synergistic anti-proliferative activity in BRCA1/2-mutated breast and ovarian cancer cells. The combination therapy also demonstrated synergistic tumor inhibition in a xenograft mouse model. Mechanistically, olaparib monotherapy increased the expressions of p-ATM and DNA-PKcs, suggesting the activation of a DNA repair pathway, whereas combining PSPC1 siRNA with olaparib decreased the expressions of p-ATM and DNA-PKcs again. As such, the combination increased the formation of γH2AX foci, indicating stronger DNA double-strand breaks. Subsequently, these DNA-damaged cells escaped G2/M checkpoint activation, as indicated by the suppression of p-cdc25C (Ser216) and p-cdc2 (Tyr15) after combination treatment. Finally, these cells entered mitosis, which induced increased apoptosis. Thus, this proves that PSPC1 inhibition enhances olaparib sensitivity by targeting DNA damage response in our preclinical model. The combination of olaparib and PSPC1 inhibition merits further clinical investigation to enhance PARP inhibitor efficacy.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Neoplasias Ováricas , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Antineoplásicos/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Humanos , Femenino , Ratones , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA2/genética , ARN Interferente Pequeño/genética
4.
Mol Carcinog ; 61(1): 73-84, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34699643

RESUMEN

Paraspeckles compound 1 (PSPC1) is a multifunctional protein that plays an important role in cancer cells, where PSPC1 is a master regulator of pro-oncogenic responses that includes activation of TGFß (TGFß1), TGFß-dependent EMT, and metastasis. The pro-oncogenic activities of PSPC1 closely resembled those observed for the orphan nuclear receptor 4A1 (NR4A1, Nur77) and knockdown of NR4A1 decreased expression of PSPC1 in MDA-MB-231 breast, H1299 lung, and SNU449 liver cancer cells. Similar results were observed in these same cell lines after treatment with bisindole-derived (CDIMs) NR4A1 antagonists. Moreover, PSPC1-dependent regulation of TGFß, genes associated with cancer stem cells and epithelial to mesenchymal transition (EMT) were also downregulated after NR4A1 silencing or treatment of breast, lung, and liver cancer cells with CDIM/NR4A1 antagonists. Results of chromatin immunoprecipitation (ChIP) assays suggest that NR4A1 regulates PSPC1 through interaction with an NBRE sequence in the PSPC1 gene promoter. These results coupled with in vivo studies showing that NR4A1 antagonists inhibit breast tumor growth and downregulate PSPC1 in tumors indicate that the pro-oncogenic nuclear PSPC1 factor can be targeted by CDIM/NR4A1 antagonists.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Metano/administración & dosificación , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Regiones Promotoras Genéticas , Proteínas de Unión al ARN/genética , Células A549 , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Células HCT116 , Células Hep G2 , Humanos , Metano/farmacología , Ratones , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/antagonistas & inhibidores , Células PC-3 , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Transl Med ; 20(1): 389, 2022 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-36059030

RESUMEN

BACKGROUND: Streptococcus pneumoniae is the leading reason for invasive diseases including pneumonia and meningitis, and also secondary infections following viral respiratory diseases such as flu and COVID-19. Currently, serotype-dependent vaccines, which have several insufficiency and limitations, are the only way to prevent pneumococcal infections. Hence, it is plain to need an alternative effective strategy for prevention of this organism. Protein-based vaccine involving conserved pneumococcal protein antigens with different roles in virulence could provide an eligible alternative to existing vaccines. METHODS: In this study, PspC, PhtD and PsaA antigens from pneumococcus were taken to account to predict B-cell and helper T-cell epitopes, and epitope-rich regions were chosen to build the construct. To enhance the immunogenicity of the epitope-based vaccine, a truncated N-terminal fragment of pneumococcal endopeptidase O (PepO) was used as a potential TLR2/4 agonist which was identified by molecular docking studies. The ultimate construct was consisted of the chosen epitope-rich regions, along with the adjuvant role (truncated N-PepO) and suitable linkers. RESULTS: The epitope-based vaccine was assessed as regards physicochemical properties, allergenicity, antigenicity, and toxicity. The 3D structure of the engineered construct was modeled, refined, and validated. Molecular docking and simulation of molecular dynamics (MD) indicated the proper and stable interactions between the vaccine and TLR2/4 throughout the simulation periods. CONCLUSIONS: For the first time this work presents a novel vaccine consisting of epitopes of PspC, PhtD, and PsaA antigens which is adjuvanted with a new truncated domain of PepO. The computational outcomes revealed that the suggested vaccine could be deemed an efficient therapeutic vaccine for S. pneumoniae; nevertheless, in vitro and in vivo examinations should be performed to prove the potency of the candidate vaccine.


Asunto(s)
COVID-19 , Streptococcus pneumoniae , Adyuvantes Inmunológicos , Antígenos Bacterianos , Proteínas Bacterianas , Biología Computacional , Epítopos de Linfocito B/química , Epítopos de Linfocito T/química , Humanos , Metaloendopeptidasas , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Receptor Toll-Like 2 , Vacunas de Subunidad/química
6.
J Biomed Sci ; 28(1): 57, 2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34340703

RESUMEN

Dysregulation of nucleocytoplasmic shuttling is commonly observed in cancers and emerging as a cancer hallmark for the development of anticancer therapeutic strategies. Despite its severe adverse effects, selinexor, a selective first-in-class inhibitor of the common nuclear export receptor XPO1, was developed to target nucleocytoplasmic protein shuttling and received accelerated FDA approval in 2019 in combination with dexamethasone as a fifth-line therapeutic option for adults with relapsed refractory multiple myeloma (RRMM). To explore innovative targets in nucleocytoplasmic shuttling, we propose that the aberrant contextual determinants of nucleocytoplasmic shuttling, such as PSPC1 (Paraspeckle component 1), TGIF1 (TGF-ß Induced Factor Homeobox 1), NPM1 (Nucleophosmin), Mortalin and EBP50, that modulate shuttling (or cargo) proteins with opposite tumorigenic functions in different subcellular locations could be theranostic targets for developing anticancer strategies. For instance, PSPC1 was recently shown to be the contextual determinant of the TGF-ß prometastatic switch and PTK6/ß-catenin reciprocal oncogenic nucleocytoplasmic shuttling during hepatocellular carcinoma (HCC) progression. The innovative nucleocytoplasmic shuttling inhibitor PSPC1 C-terminal 131 polypeptide (PSPC1-CT131), which was developed to target both the shuttling determinant PSPC1 and the shuttling protein PTK6, maintained their tumor-suppressive characteristics and exhibited synergistic effects on tumor suppression in HCC cells and mouse models. In summary, targeting the contextual determinants of nucleocytoplasmic shuttling with cargo proteins having opposite tumorigenic functions in different subcellular locations could be an innovative strategy for developing new therapeutic biomarkers and agents to improve cancer therapy.


Asunto(s)
Progresión de la Enfermedad , Neoplasias/genética , Oncogenes , Proteínas de Unión al ARN/genética , Translocación Genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Nucleofosmina , Proteínas de Unión al ARN/metabolismo
7.
Acta Biochim Biophys Sin (Shanghai) ; 53(11): 1527-1537, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34490876

RESUMEN

Liquid-liquid phase separation (LLPS) underlies the formation of membraneless compartments in mammal cells. However, there are few reports that focus on the correlation of mouse oocyte maturation with LLPS. Previous studies have reported that paraspeckle component 1 (PSPC1) is related to the occurrence and development of tumors, but whether PSPC1 functions in mouse oocyte maturation is still unclear. Sequence analysis of PSPC1 protein showed that it contains a prion-like domain (PrLD) that is required for phase separation of proteins. In this study, we found that PSPC1 could undergo phase separation. Moreover, the loss of PrLD domain of PSPC1 could greatly weaken its phase separation ability. The immunofluorescence assays showed that PSPC1 is present in mouse oocytes in the germinal vesicle (GV) stage. Knockdown of PSPC1 significantly impeded the maturation of mouse oocytes in vitro. CHK1 has been reported to play important roles in the GV stage of mouse oocytes. Co-IP experiment revealed that PSPC1 could interact with phosphatase serine/threonine-protein phosphatase 5 (PPP5C), which regulates CHK1 phosphorylation. Western blot analysis revealed that PSPC1 could regulate the phosphorylation of CHK1 through PPP5C; however, PSPC1 without PrLD domain was inactive, suggesting that the lack of phase separation ability led to the abnormal function of PSPC1 in regulating CHK1 phosphorylation. Thus, we conclude that PSPC1 may undergo phase separation to regulate the phosphorylation level of CHK1 via PPP5C and participate in mouse oocyte maturation. Our study provides new insights into the mechanism of mouse oocyte maturation.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Proteínas Nucleares/genética , Oocitos/metabolismo , Fosfoproteínas Fosfatasas/genética , Procesamiento Proteico-Postraduccional , Proteínas de Unión al ARN/genética , Secuencia de Aminoácidos , Animales , Diferenciación Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Hep G2 , Humanos , Ratones , Ratones Endogámicos ICR , Proteínas Nucleares/metabolismo , Oocitos/citología , Oocitos/crecimiento & desarrollo , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Transducción de Señal
8.
J Proteome Res ; 19(4): 1824-1846, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32108472

RESUMEN

Lactobacillus casei group bacteria improve cheese ripening and may interact with host intestinal cells as probiotics, where surface proteins play a key role. Three complementary methods [trypsin shaving (TS), LiCl-sucrose (LS) extraction, and extracellular culture fluid precipitation] were used to analyze cell surface proteins of Lactobacillus paracasei GCRL163 by label-free quantitative proteomics after culture to the mid-exponential phase in bioreactors at pH 6.5 and temperatures of 30-45 °C. A total of 416 proteins, including 300 with transmembrane, cell wall anchoring, and secretory motifs and 116 cytoplasmic proteins, were quantified as surface proteins. Although LS caused significantly greater cell lysis as growth temperature increased, higher numbers of extracytoplasmic proteins were exclusively obtained by LS treatment. Together with the increased positive surface charge of cells cultured at supra-optimal temperatures, proteins including cell wall hydrolases Msp1/p75 and Msp2/p40, α-fucosidase AlfB, SecA, and a PspC-domain putative adhesin were upregulated in surface or secreted protein fractions, suggesting that cell adhesion may be altered. Prolonged heat stress (PHS) increased binding of L. paracasei GCRL163 to human colorectal adenocarcinoma HT-29 cells, relative to acid-stressed cells. This study demonstrates that PHS influences cell adhesion and relative abundance of proteins located on the surface, which may impact probiotic functionality, and the detected novel surface proteins likely linked to the cell cycle and envelope stress.


Asunto(s)
Adenocarcinoma , Neoplasias Colorrectales , Lacticaseibacillus paracasei , Probióticos , Neoplasias Colorrectales/genética , Células HT29 , Respuesta al Choque Térmico , Humanos , Proteínas de la Membrana/genética
9.
J Biol Chem ; 293(17): 6593-6602, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29530979

RESUMEN

Members of the Drosophila behavior human splicing (DBHS) protein family are nuclear proteins implicated in many layers of nuclear functions, including RNA biogenesis as well as DNA repair. Definitive of the DBHS protein family, the conserved DBHS domain provides a dimerization platform that is critical for the structural integrity and function of these proteins. The three human DBHS proteins, splicing factor proline- and glutamine-rich (SFPQ), paraspeckle component 1 (PSPC1), and non-POU domain-containing octamer-binding protein (NONO), form either homo- or heterodimers; however, the relative affinity and mechanistic details of preferential heterodimerization are yet to be deciphered. Here we report the crystal structure of a SFPQ/PSPC1 heterodimer to 2.3-Å resolution and analyzed the subtle structural differences between the SFPQ/PSPC1 heterodimer and the previously characterized SFPQ homodimer. Analytical ultracentrifugation to estimate the dimerization equilibrium of the SFPQ-containing dimers revealed that the SFPQ-containing dimers dissociate at low micromolar concentrations and that the heterodimers have higher affinities than the homodimer. Moreover, we observed that the apparent dissociation constant for the SFPQ/PSPC1 heterodimer was over 6-fold lower than that of the SFPQ/NONO heterodimer. We propose that these differences in dimerization affinity may represent a potential mechanism by which PSPC1 at a lower relative cellular abundance can outcompete NONO to heterodimerize with SFPQ.


Asunto(s)
Proteínas Nucleares/química , Factor de Empalme Asociado a PTB/química , Multimerización de Proteína , Proteínas de Unión al ARN/química , Cristalografía por Rayos X , Humanos , Proteínas Nucleares/metabolismo , Factor de Empalme Asociado a PTB/metabolismo , Estructura Cuaternaria de Proteína , Proteínas de Unión al ARN/metabolismo
10.
J Infect Dis ; 217(3): 358-370, 2018 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28968817

RESUMEN

Pneumococcal hemolytic uremic syndrome (HUS) in children is caused by infections with Streptococcus pneumoniae. Because endothelial cell damage is a hallmark of HUS, we studied how HUS-inducing pneumococci derived from infant HUS patients during the acute phase disrupt the endothelial layer. HUS pneumococci efficiently bound human plasminogen. These clinical isolates of HUS pneumococci efficiently bound human plasminogen via the bacterial surface proteins Tuf and PspC. When activated to plasmin at the bacterial surface, the active protease degraded fibrinogen and cleaved C3b. Here, we show that PspC is a pneumococcal plasminogen receptor and that plasmin generated on the surface of HUS pneumococci damages endothelial cells, causing endothelial retraction and exposure of the underlying matrix. Thus, HUS pneumococci damage endothelial cells in the blood vessels and disturb local complement homeostasis. Thereby, HUS pneumococci promote a thrombogenic state that drives HUS pathology.


Asunto(s)
Adhesión Bacteriana , Proteínas Bacterianas/metabolismo , Células Endoteliales/patología , Fibrinolisina/metabolismo , Síndrome Hemolítico-Urémico/microbiología , Plasminógeno/metabolismo , Streptococcus pneumoniae/fisiología , Preescolar , Femenino , Humanos , Infecciones Neumocócicas/microbiología , Unión Proteica , Streptococcus pneumoniae/aislamiento & purificación
11.
Infect Immun ; 86(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29378798

RESUMEN

The pneumococcal capsular serotype is an important determinant of complement resistance and invasive disease potential, but other virulence factors have also been found to contribute. Pneumococcal surface protein C (PspC), a highly variable virulence protein that binds complement factor H to evade C3 opsonization, is divided into two subgroups: choline-bound subgroup I and LPxTG-anchored subgroup II. The prevalence of different PspC subgroups in invasive pneumococcal disease (IPD) and functional differences in complement evasion are unknown. The prevalence of PspC subgroups in IPD isolates was determined in a collection of 349 sequenced strains of Streptococcus pneumoniae isolated from adult patients. pspC deletion mutants and isogenic pspC switch mutants were constructed to study differences in factor H binding and complement evasion in relation to capsule thickness. Subgroup I pspC was far more prevalent in IPD isolates than subgroup II pspC The presence of capsule was associated with a greater ability of bound factor H to reduce complement opsonization. Pneumococcal subgroup I PspC bound significantly more factor H and showed more effective complement evasion than subgroup II PspC in isogenic encapsulated pneumococci. We conclude that variation in the PspC subgroups, independent of capsule serotypes, affects pneumococcal factor H binding and its ability to evade complement deposition.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Proteínas del Sistema Complemento/inmunología , Genotipo , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología , Anciano , Factor H de Complemento/inmunología , Factor H de Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Femenino , Humanos , Evasión Inmune , Masculino , Persona de Mediana Edad , Tipificación Molecular , Mutación , Infecciones Neumocócicas/epidemiología , Prevalencia , Serogrupo , Virulencia/genética , Factores de Virulencia/genética
12.
J Infect Dis ; 215(5): 713-722, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28035010

RESUMEN

Background: Several Streptococcus pneumoniae proteins play a role in pathogenesis and are being investigated as vaccine targets. It is largely unknown whether naturally acquired antibodies reduce the risk of colonization with strains expressing a particular antigenic variant. Methods: Serum immunoglobulin G (IgG) titers to 28 pneumococcal protein antigens were measured among 242 individuals aged <6 months-78 years in Native American communities between 2007 and 2009. Nasopharyngeal swabs were collected >- 30 days after serum collection, and the antigen variant in each pneumococcal isolate was determined using genomic data. We assessed the association between preexisting variant-specific antibody titers and subsequent carriage of pneumococcus expressing a particular antigen variant. Results: Antibody titers often increased across pediatric groups before decreasing among adults. Individuals with low titers against group 3 pneumococcal surface protein C (PspC) variants were more likely to be colonized with pneumococci expressing those variants. For other antigens, variant-specific IgG titers do not predict colonization. Conclusion: We observed an inverse association between variant-specific antibody concentration and homologous pneumococcal colonization for only 1 protein. Further assessment of antibody repertoires may elucidate the nature of antipneumococcal antibody-mediated mucosal immunity while informing vaccine development.


Asunto(s)
Factores de Edad , Anticuerpos Antibacterianos/sangre , Antígenos Bacterianos/sangre , Infecciones Neumocócicas/sangre , Adolescente , Adulto , Anciano , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/sangre , Portador Sano/inmunología , Portador Sano/microbiología , Niño , Preescolar , Estudios de Seguimiento , Proteínas de Choque Térmico/sangre , Humanos , Inmunoglobulina G/sangre , Lactante , Modelos Logísticos , Estudios Longitudinales , Persona de Mediana Edad , Nasofaringe/microbiología , Infecciones Neumocócicas/inmunología , Estudios Prospectivos , Adulto Joven
13.
J Infect Dis ; 209(3): 377-88, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24009156

RESUMEN

BACKGROUND: Pneumococcal serotypes are represented by a varying number of clonal lineages with different genetic contents, potentially affecting invasiveness. However, genetic variation within the same genetic lineage may be larger than anticipated. METHODS: A total of 715 invasive and carriage isolates from children in the same region and during the same period were compared using pulsed-field gel electrophoresis (PFGE) and multilocus sequence typing. Bacterial genome sequencing, functional assays, and in vivo virulence mice studies were performed. RESULTS: Clonal types of the same serotype but also intraclonal variants within clonal complexes (CCs) showed differences in invasive-disease potential. CC138, a common CC, was divided into several PFGE patterns, partly explained by number, location, and type of temperate bacteriophages. Whole-genome sequencing of 4 CC138 isolates representing PFGE clones with different invasive-disease potentials revealed intraclonal sequence variations of the virulence-associated proteins pneumococcal surface protein A (PspA) and pneumococcal choline-binding protein C (PspC). A carrier isolate lacking PcpA exhibited decreased virulence in mice, and there was a differential binding of human factor H, depending on invasiveness. CONCLUSIONS: Pneumococcal clonal types but also intraclonal variants exhibited different invasive-disease potentials in children. Intraclonal variants, reflecting different prophage contents, showed differences in major surface antigens. This suggests ongoing immune selection, such as that due to PspC-mediated complement resistance through varied human factor H binding, that may affect invasiveness in children.


Asunto(s)
Variación Genética , Infecciones Neumocócicas/epidemiología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/genética , Adolescente , Animales , Antígenos Bacterianos/análisis , Portador Sano/epidemiología , Portador Sano/microbiología , Niño , Preescolar , Modelos Animales de Enfermedad , Electroforesis en Gel de Campo Pulsado , Femenino , Genoma Bacteriano , Genotipo , Humanos , Lactante , Masculino , Proteínas de la Membrana/análisis , Ratones , Ratones Endogámicos C57BL , Tipificación Molecular , Infecciones Neumocócicas/microbiología , Profagos/genética , Análisis de Secuencia de ADN , Fagos de Streptococcus/genética , Streptococcus pneumoniae/aislamiento & purificación , Virulencia
14.
J Biol Chem ; 288(22): 15614-27, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23603906

RESUMEN

Adherence of Streptococcus pneumoniae is directly mediated by interactions of adhesins with eukaryotic cellular receptors or indirectly by exploiting matrix and serum proteins as molecular bridges. Pneumococci engage vitronectin, the human adhesive glycoprotein and complement inhibitor, to facilitate attachment to epithelial cells of the mucosal cavity, thereby modulating host cell signaling. In this study, we identified PspC as a vitronectin-binding protein interacting with the C-terminal heparin-binding domain of vitronectin. PspC is a multifunctional surface-exposed choline-binding protein displaying various adhesive properties. Vitronectin binding required the R domains in the mature PspC protein, which are also essential for the interaction with the ectodomain of the polymeric immunoglobulin receptor and secretory IgA. Consequently, secretory IgA competitively inhibited binding of vitronectin to purified PspC and to PspC-expressing pneumococci. In contrast, Factor H, which binds to the N-terminal part of mature PspC molecules, did not interfere with the PspC-vitronectin interaction. Using a series of vitronectin peptides, the C-terminal heparin-binding domain was shown to be essential for the interaction of soluble vitronectin with PspC. Binding experiments with immobilized vitronectin suggested a region N-terminal to the identified heparin-binding domain as an additional binding region for PspC, suggesting that soluble, immobilized, as well as cellularly bound vitronectin possesses different conformations. Finally, vitronectin bound to PspC was functionally active and inhibited the deposition of the terminal complement complex. In conclusion, this study identifies and characterizes (on the molecular level) the interaction between the pneumococcal adhesin PspC and the human glycoprotein vitronectin.


Asunto(s)
Proteínas Bacterianas/metabolismo , Streptococcus pneumoniae/metabolismo , Vitronectina/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Humanos , Unión Proteica , Estructura Terciaria de Proteína , Streptococcus pneumoniae/química , Streptococcus pneumoniae/genética , Vitronectina/química , Vitronectina/genética
15.
Int J Med Microbiol ; 304(8): 1233-46, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25455218

RESUMEN

Colonization of Streptococcus pneumoniae (pneumococci) is a prerequisite for bacterial dissemination and their capability to enter the bloodstream. Pneumococci have evolved various successful strategies to colonize the mucosal epithelial barrier of humans. A pivotal mechanism of host cell invasion implicated with invasive diseases is promoted by the interaction of pneumococcal PspC with the polymeric Ig-receptor (pIgR). However, the mechanism(s) of pneumococcal endocytosis and the intracellular route of pneumococci upon uptake by the PspC-pIgR-interaction are not known. Here, we demonstrate by using a combination of pharmacological inhibitors and genetics interference approaches the involvement of active dynamin-dependent caveolae and clathrin-coated vesicles for pneumococcal uptake via the PspC-pIgR mechanism. Depleting cholesterol from host cell membranes and disruption of lipid microdomains impaired pneumococcal internalization. Moreover, chemical inhibition of clathrin or functional inactivation of dynamin, caveolae or clathrin by RNA interference significantly affected pneumococcal internalization suggesting that clathrin-mediated endocytosis (CME) and caveolae are involved in the bacterial uptake process. Confocal fluorescence microscopy of pIgR-expressing epithelial cells infected with pneumococci or heterologous Lactococcus lactis expressing PspC demonstrated bacterial co-localization with fluorescent-tagged clathrin and early as well as recycling or late endosomal markers such as Lamp1, Rab5, Rab4, and Rab7, respectively. In conclusion these data suggest that PspC-promoted uptake is mediated by both CME and caveolae. After endocytosis pneumococci are routed via the endocytic pathway into early endosomes and are then sorted into recycling or late endosomes, which can result in pneumococcal killing in phagolysosomes or transcytosis via recycling endosomes.


Asunto(s)
Adhesión Bacteriana , Caveolinas/metabolismo , Clatrina/metabolismo , Endocitosis , Células Epiteliales/fisiología , Receptores de Inmunoglobulina Polimérica/metabolismo , Streptococcus pneumoniae/fisiología , Animales , Proteínas Bacterianas/metabolismo , Línea Celular , Perros , Células Epiteliales/microbiología , Humanos , Unión Proteica
16.
Cancer Lett ; 587: 216733, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38360141

RESUMEN

Despite significant advances in diagnostic techniques and treatment approaches, the prognosis of pancreatic ductal adenocarcinoma (PDAC) is still poor. Previous studies have reported that S-phase kinase-associated protein 2 (SKP2), a subunit of the SCF E3 ubiquitin ligase complex, is engaged in the malignant biological behavior of some tumor entities. However, SKP2 has not been fully investigated in PDAC. In the present study, it was observed that high expression of SKP2 significantly correlates with decreased survival time. Further experiments suggested that SKP2 promotes metastasis by interacting with the putative transcription factor paraspeckle component 1 (PSPC1). According to the results of coimmunoprecipitation and ubiquitination assays, SKP2 depletion resulted in the polyubiquitination of PSPC1, followed by its degradation. Furthermore, the SKP2-mediated ubiquitination of PSPC1 partially depended on the activity of the E3 ligase TRIM21. In addition, inhibition of the SKP2/PSPC1 axis by SMIP004, a traditional inhibitor of SKP2, impaired the migration of PDAC cells. In summary, this study provides novel insight into the mechanisms involved in PDAC malignant progression. Targeting the SKP2/PSPC1 axis is a promising strategy for the treatment of PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Proteínas Quinasas Asociadas a Fase-S/genética , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Neoplasias Pancreáticas/genética , Ubiquitinación , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Carcinoma Ductal Pancreático/genética , Proteínas de Unión al ARN/metabolismo
17.
Viruses ; 16(5)2024 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-38793620

RESUMEN

Hepatitis C virus (HCV) infects the human liver, and its chronic infection is one of the major causes of Hepatocellular carcinoma. Translation of HCV RNA is mediated by an Internal Ribosome Entry Site (IRES) element located in the 5'UTR of viral RNA. Several RNA Binding proteins of the host interact with the HCV IRES and modulate its function. Here, we demonstrate that PSPC1 (Paraspeckle Component 1), an essential paraspeckle component, upon HCV infection is relocalized and interacts with HCV IRES to prevent viral RNA translation. Competition UV-crosslinking experiments showed that PSPC1 interacts explicitly with the SLIV region of the HCV IRES, which is known to play a vital role in ribosomal loading to the HCV IRES via interaction with Ribosomal protein S5 (RPS5). Partial silencing of PSPC1 increased viral RNA translation and, consequently, HCV replication, suggesting a negative regulation by PSPC1. Interestingly, the silencing of PSPC1 protein leads to an increased interaction of RPS5 at the SLIV region, leading to an overall increase in the viral RNA in polysomes. Overall, our results showed how the host counters viral infection by relocalizing nuclear protein to the cytoplasm as a survival strategy.


Asunto(s)
Hepacivirus , Sitios Internos de Entrada al Ribosoma , Biosíntesis de Proteínas , ARN Viral , Proteínas de Unión al ARN , Proteínas Ribosómicas , Replicación Viral , Hepacivirus/genética , Hepacivirus/fisiología , Humanos , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/genética , ARN Viral/metabolismo , ARN Viral/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Unión Proteica , Hepatitis C/virología , Hepatitis C/metabolismo , Interacciones Huésped-Patógeno
18.
Int J Biol Macromol ; 258(Pt 2): 128924, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38143051

RESUMEN

Pneumococcus is the top cause of diseases such as pneumonia/meningitis, and of secondary infections after viral respiratory diseases like COVID-19/flu. Pneumococcal protein-based vaccines consisting of proteins with various functions in virulence might provide a qualified alternative for present vaccines. In this project, PspC, PsaA, and PhtD proteins were considered to anticipate B/T-cell epitopes using immunoinformatics to develop 4 multi-peptide constructs (C, A, and D individual constructs, and a fusion construct CAD). We tested whether vaccination with CAD is able to elicit more efficient protective responses against infection than vaccination with the individual constructs or combination of C + A + D. Based on the in silico results, the constructs were predicted to be antigenic, soluble, non-toxic, and stable, and also be able to provoke humoral/cellular immune reactions. When mice were immunized with the fusion protein, significantly higher levels of IgG and cytokines were induced in serum. The IgG in the fusion group had an effective bioactivity for pneumococcus clearance utilizing the complement pathway. The mice immunized with fusion protein were the most protected from challenge. This report for the first time presents a novel multi-peptide vaccine composed of immunodominant peptides of PspC, PsaA, and PhtD. In general, the experimental results supported the immunoinformatics predictions.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Animales , Ratones , Proteínas Bacterianas , Vacunas de Subunidades Proteicas , Péptidos , Epítopos de Linfocito B , Inmunoglobulina G , Anticuerpos Antibacterianos
19.
Clin Transl Oncol ; 25(3): 662-672, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36422798

RESUMEN

PURPOSE: Aberrant activation of STAT3 signal pathway promotes tumor progression in many solid tumor types, including cervical cancer and endometrial cancer. BBI608, the STAT3 inhibitor had been reported in previous studies for restraining cancer stem cells. However, whether BBI608 is available for inhibiting the proliferation of cervical cancer or endometrial cancer remains poorly understood. This study investigated the anti-tumor effect and molecular mechanism of BBI608 on the patient-specific primary cells (PSPC) generated from cervical and endometrial cancer in vitro. METHODS: PSPCs were obtained from four patients via biopsy. The cell viability was analyzed by the CCK8 assay. The PSPCs were treated with various concentrations of BBI608 or/and paclitaxel; and then, western blot was applied to investigate the expression of phosphorylated STAT3 (pSTAT3). RESULTS: The PSPCs cell viability was reduced after treated with BBI608 at a lower concentration. Western blot results showed a reduction trend of pSTAT3 after PSPCs treated with BBI608. Our results demonstrated that BBI608 at the certain concentrations worked well in reducing the cell viability of PSPC from the patients who suffered from cervical cancer and endometrial cancer. CONCLUSIONS: In this study, the patient-specific primary cell (PSPC) was used as the pre-clinical model for investigating the efficiency of BBI608 in reducing cancer cells viability. BBI608, at a clinical-relevant concentration, had valid efficiency in PSPCs from the patients. The dose of drugs treatment and the measured results were more valuable for further guiding clinical trials.


Asunto(s)
Neoplasias Endometriales , Neoplasias del Cuello Uterino , Femenino , Humanos , Neoplasias del Cuello Uterino/tratamiento farmacológico , Supervivencia Celular , Neoplasias Endometriales/patología , Paclitaxel/farmacología , Factor de Transcripción STAT3/metabolismo , Línea Celular Tumoral , Proliferación Celular
20.
Hum Pathol ; 138: 1-11, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37209920

RESUMEN

Prostate cancer (PCa) remains the most commonly diagnosed cancer in men worldwide and is still the second leading cause of cancer-related death. One major cause of PCa development is epigenetic aberration, including histone modification. We have previously demonstrated that Lysine Demethylase 5C (KDM5C) plays an essential role in the development of PCa and drives PCa progression by promoting epithelial-mesenchymal transition. Epigenetic regulators often work in concert, for example, to regulate transcription. We identified Paraspeckle Component 1 (PSPC1) as an interacting protein of KDM5C, suggesting that these proteins might function together in PCa. Here, we systematically investigate the expression patterns of KDM5C and PSPC1 in 2 independent prostate cohorts (432 and 205 prostate tumors in total for PSPC1 and KDM5C, respectively) by immunohistochemistry. We demonstrate that the expression of PSPC1 correlates with that of KDM5C. In addition, PSPC1 is up-regulated in primary and metastatic PCa. Elevated PSPC1 expression correlates with a higher-grade group and an advanced T-stage. Patients with high PSPC1 expression have a worse biochemical recurrence-free survival. In addition, PSPC1 expression is an independent prognostic parameter. Our data indicate that KDM5C and PSPC1 are involved in PCa progression, and therapeutic inhibition of KDM5C and PSPC1 by selective compounds might be a promising approach for the treatment of PCa.


Asunto(s)
Neoplasias de la Próstata , Masculino , Humanos , Próstata , Transición Epitelial-Mesenquimal , Proteínas de Unión al ARN , Histona Demetilasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA