Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Immunol ; 206(9): 2135-2145, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33858961

RESUMEN

Asplenia imparts susceptibility to life-threatening sepsis with encapsulated bacteria, such as the pneumococcus. However, the cellular components within the splenic environment that guard against pneumococcal bacteremia have not been defined. The actin-bundling protein L-plastin (LPL) is essential for the generation of marginal zone B cells and for anti-pneumococcal host defense, as revealed by a mouse model of genetic LPL deficiency. In independent studies, serine phosphorylation of LPL at residue 5 (S5) has been described as a key "switch" in regulating LPL actin binding and subsequent cell motility, although much of the data are correlative. To test the importance of S5 phosphorylation in LPL function, and to specifically assess the requirement of LPL S5 phosphorylation in anti-pneumococcal host defense, we generated the "S5A" mouse, expressing endogenous LPL bearing a serine-to-alanine mutation at this position. S5A mice were bred to homozygosity, and LPL was expressed at levels equivalent to wild-type, but S5 phosphorylation was absent. S5A mice exhibited specific impairment in clearance of pneumococci following i.v. challenge, with 10-fold-higher bacterial bloodstream burden 24 h after challenge compared with wild-type or fully LPL-deficient animals. Defective bloodstream clearance correlated with diminished population of marginal zone macrophages and with reduced phagocytic capacity of multiple innate immune cells. Development and function of other tested leukocyte lineages, such as T and B cell motility and activation, were normal in S5A mice. The S5A mouse thus provides a novel system in which to elucidate the precise molecular control of critical immune cell functions in specific host-pathogen defense interactions.


Asunto(s)
Glicoproteínas de Membrana/inmunología , Proteínas de Microfilamentos/inmunología , Serina/inmunología , Bazo/inmunología , Streptococcus pneumoniae/inmunología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Streptococcus pneumoniae/aislamiento & purificación
2.
J Cell Biochem ; 119(12): 10351-10357, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30136304

RESUMEN

Mice with disruption of Lrrk1 and patients with nonfunctional mutant Lrrk1 exhibit severe osteopetrosis phenotypes because of osteoclast cytoskeletal dysfunction. To understand how Lrrk1 regulates osteoclast function by modulating cytoskeleton rearrangement, we examined the proteins that are differentially phosphorylated in wild-type mice and Lrrk1-deficient osteoclasts by metal affinity purification coupled liquid chromatography/mass spectrometry (LC/MS) analyses. One of the candidates that we identified by LC/MS is L-plastin, an actin bundling protein. We found that phosphorylation of L-plastin at serine (Ser) residues 5 was present in wild-type osteoclasts but not in Lrrk1-deficient cells. Western blot analyses with antibodies specific for Ser5 phosphorylated L-plastin confirmed the reduced L-plastin Ser5 phosphorylation in Lrrk1 knockout (KO) osteoclasts. micro computed tomography (Micro-CT) analyses revealed that the trabecular bone volume of the distal femur was increased by 27% in the 16 to 21-week-old L-plastin KO females as compared with the wild-type control mice. The ratio of bone volume to tissue volume and connectivity density were increased by 44% and 47% (both P < 0.05), respectively, in L-plastin KO mice. Our data suggest that targeted disruption of L-plastin increases trabecular bone volume, and phosphorylation of Ser5 in L-plastin in the Lrrk1 signaling pathway may in part contribute to actin assembly in mature osteoclasts.


Asunto(s)
Actinas/genética , Glicoproteínas de Membrana/genética , Proteínas de Microfilamentos/genética , Osteopetrosis/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Hueso Esponjoso/crecimiento & desarrollo , Hueso Esponjoso/metabolismo , Citoesqueleto/genética , Humanos , Ratones , Ratones Noqueados , Osteoclastos/metabolismo , Osteoclastos/patología , Osteopetrosis/patología , Fosforilación/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/deficiencia , Serina/genética , Transducción de Señal/genética
3.
Blood ; 128(24): 2785-2796, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27758872

RESUMEN

Alveolar macrophages are lung-resident sentinel cells that develop perinatally and protect against pulmonary infection. Molecular mechanisms controlling alveolar macrophage generation have not been fully defined. Here, we show that the actin-bundling protein L-plastin (LPL) is required for the perinatal development of alveolar macrophages. Mice expressing a conditional allele of LPL (CD11c.Crepos-LPLfl/fl) exhibited significant reductions in alveolar macrophages and failed to effectively clear pulmonary pneumococcal infection, showing that immunodeficiency results from reduced alveolar macrophage numbers. We next identified the phase of alveolar macrophage development requiring LPL. In mice, fetal monocytes arrive in the lungs during a late fetal stage, maturing to alveolar macrophages through a prealveolar macrophage intermediate. LPL was required for the transition from prealveolar macrophages to mature alveolar macrophages. The transition from prealveolar macrophage to alveolar macrophage requires the upregulation of the transcription factor peroxisome proliferator-activated receptor-γ (PPAR-γ), which is induced by exposure to granulocyte-macrophage colony-stimulating factor (GM-CSF). Despite abundant lung GM-CSF and intact GM-CSF receptor signaling, PPAR-γ was not sufficiently upregulated in developing alveolar macrophages in LPL-/- pups, suggesting that precursor cells were not correctly localized to the alveoli, where GM-CSF is produced. We found that LPL supports 2 actin-based processes essential for correct localization of alveolar macrophage precursors: (1) transmigration into the alveoli, and (2) engraftment in the alveoli. We thus identify a molecular pathway governing neonatal alveolar macrophage development and show that genetic disruption of alveolar macrophage development results in immunodeficiency.


Asunto(s)
Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Animales , Animales Recién Nacidos , Antígenos CD11/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Ratones Endogámicos C57BL , Modelos Biológicos , Monocitos/metabolismo , PPAR gamma/metabolismo , Infecciones Neumocócicas/patología , Podosomas/metabolismo , Transporte de Proteínas , Regulación hacia Arriba/efectos de los fármacos
4.
J Immunol ; 197(5): 1683-91, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27465533

RESUMEN

Exploring the mechanisms controlling lymphocyte trafficking is essential for understanding the function of the immune system and the pathophysiology of immunodeficiencies. The mammalian Ste20-like kinase 1 (Mst1) has been identified as a critical signaling mediator of T cell migration, and loss of Mst1 results in immunodeficiency disease. Although Mst1 is known to support T cell migration through induction of cell polarization and lamellipodial formation, the downstream effectors of Mst1 are incompletely defined. Mice deficient for the actin-bundling protein L-plastin (LPL) have phenotypes similar to mice lacking Mst1, including decreased T cell polarization, lamellipodial formation, and cell migration. We therefore asked whether LPL functions downstream of Mst1. The regulatory N-terminal domain of LPL contains a consensus Mst1 phosphorylation site at Thr(89) We found that Mst1 can phosphorylate LPL in vitro and that Mst1 can interact with LPL in cells. Removal of the Mst1 phosphorylation site by mutating Thr(89) to Ala impaired localization of LPL to the actin-rich lamellipodia of T cells. Expression of the T89A LPL mutant failed to restore migration of LPL-deficient T cells in vitro. Furthermore, expression of T89A LPL in LPL-deficient hematopoietic cells, using bone marrow chimeras, failed to rescue the phenotype of decreased thymic egress. These results identify LPL as a key effector of Mst1 and establish a novel mechanism linking a signaling intermediate to an actin-binding protein critical to T cell migration.


Asunto(s)
Movimiento Celular , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Animales , Proteínas del Citoesqueleto , Citometría de Flujo , Activación de Linfocitos , Linfocitos/inmunología , Ratones , Proteínas de Microfilamentos , Fosfoproteínas/deficiencia , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Seudópodos/inmunología , Seudópodos/fisiología
5.
J Infect Dis ; 213(4): 649-58, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26347570

RESUMEN

Klebsiella pneumoniae, a chief cause of nosocomial pneumonia, is a versatile and commonly multidrug-resistant human pathogen for which further insight into pathogenesis is needed. We show that the pilus regulatory gene fimK promotes the virulence of K. pneumoniae strain TOP52 in murine pneumonia. This contrasts with the attenuating effect of fimK on urinary tract virulence, illustrating that a single factor may exert opposing effects on pathogenesis in distinct host niches. Loss of fimK in TOP52 pneumonia was associated with diminished lung bacterial burden, limited innate responses within the lung, and improved host survival. FimK expression was shown to promote serum resistance, capsule production, and protection from phagocytosis by host immune cells. Finally, while the widely used K. pneumoniae model strain 43816 produces rapid dissemination and death in mice, TOP52 caused largely localized pneumonia with limited lethality, thereby providing an alternative tool for studying K. pneumoniae pathogenesis and control within the lung.


Asunto(s)
Klebsiella pneumoniae/crecimiento & desarrollo , Neumonía Bacteriana/microbiología , Factores de Virulencia/metabolismo , Animales , Cápsulas Bacterianas/inmunología , Cápsulas Bacterianas/metabolismo , Carga Bacteriana , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Humanos , Inmunidad Innata , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/inmunología , Pulmón/microbiología , Ratones Endogámicos C57BL , Fagocitosis , Neumonía Bacteriana/inmunología , Análisis de Supervivencia , Virulencia , Factores de Virulencia/genética
6.
Infect Immun ; 82(5): 1982-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24595139

RESUMEN

We report that mice deficient for the hematopoietic-specific, actin-bundling protein L-plastin (LPL) succumb rapidly to intratracheal pneumococcal infection. The increased susceptibility of LPL(-/-) mice to pulmonary pneumococcal challenge correlated with reduced numbers of alveolar macrophages, consistent with a critical role for this cell type in the immediate response to pneumococcal infection. LPL(-/-) mice demonstrated a very early clearance defect, with an almost 10-fold-higher bacterial burden in the bronchoalveolar lavage fluid 3 h following infection. Clearance of pneumococci from the alveolar space in LPL(-/-) mice was defective compared to that in Rag1(-/-) mice, which lack all B and T lymphocytes, indicating that innate immunity is defective in LPL(-/-) mice. We did not identify defects in neutrophil or monocyte recruitment or in the production of inflammatory cytokines or chemokines that would explain the early clearance defect. However, efficient alveolar macrophage regeneration following irradiation required LPL. We thus identify LPL as being key to alveolar macrophage development and essential to an effective antipneumococcal response. Further analysis of LPL(-/-) mice will illuminate critical regulators of the generation of alveolar macrophages and, thus, effective pulmonary innate immunity.


Asunto(s)
Macrófagos Alveolares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Neumonía Neumocócica/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Proliferación Celular , Femenino , Regulación de la Expresión Génica/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Eur J Immunol ; 43(7): 1735-44, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23589339

RESUMEN

Germinal center development, critical for long-term humoral immunity, requires the trafficking of T and B lymphocytes to defined tissues and locations after antigenic challenge. The molecular mechanisms that support lymphocyte trafficking through the linkage of extracellular chemotactic and adhesive cues to the actin cytoskeleton are not yet fully defined. We have previously identified the actin-bundling protein L-plastin (LPL) as a requisite intermediary in both naive B and T lymphocyte migration and in T-cell activation. We tested the hypothesis that humoral immunity would require LPL. We show that mice lacking LPL demonstrated defective germinal center formation and reduced production of T-cell-dependent antibodies. T cells from LPL(-/-) mice exhibited defective expansion of the follicular helper T population. Reduced expansion of LPL(-/-) follicular helper T cells correlated with impaired trafficking to or retention of cells in the spleen following challenge, highlighting the importance of initial lymphocyte recruitment to the eventual success of the immune response. Furthermore, LPL(-/-) B cells demonstrated cell-intrinsic defects in population expansion and in differentiation into germinal center B cells. LPL thus modulates both T- and B-cell function during the germinal center reaction and the production of T-cell-dependent antibody responses.


Asunto(s)
Formación de Anticuerpos/inmunología , Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Fosfoproteínas/inmunología , Linfocitos T/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Proteínas del Citoesqueleto , Citometría de Flujo , Centro Germinal/inmunología , Inmunidad Humoral/inmunología , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos
8.
J Immunol ; 187(6): 3015-25, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21832165

RESUMEN

B cell development is exquisitely sensitive to location within specialized niches in the bone marrow and spleen. Location within these niches is carefully orchestrated through chemotactic and adhesive cues. In this article, we demonstrate the requirement for the actin-bundling protein L-plastin (LPL) in B cell motility toward the chemokines CXCL12 and CXCL13 and the lipid chemoattractant sphingosine-1-phosphate, which guide normal B cell development. Impaired motility of B cells in LPL(-/-) mice correlated with diminished splenic maturation of B cells, with a moderate (40%) loss of follicular B cells and a profound (>80%) loss of marginal zone B cells. Entry of LPL(-/-) B cells into the lymph nodes and bone marrow of mice was also impaired. Furthermore, LPL was required for the integrin-mediated enhancement of Transwell migration but was dispensable for integrin-mediated lymphocyte adhesion. These results suggest that LPL may participate in signaling that enables lymphocyte transmigration. In support of this hypothesis, the phosphorylation of Pyk-2, a tyrosine kinase that integrates chemotactic and adhesive cues, is diminished in LPL(-/-) B cells stimulated with chemokine. Finally, a well-characterized role of marginal zone B cells is the generation of a rapid humoral response to polysaccharide Ags. LPL(-/-) mice exhibited a defective Ab response to Streptococcus pneumoniae, indicating a functional consequence of defective marginal zone B cell development in LPL(-/-) mice.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular/inmunología , Quimiotaxis de Leucocito/inmunología , Fosfoproteínas/inmunología , Actinas/metabolismo , Animales , Linfocitos B/inmunología , Adhesión Celular/inmunología , Separación Celular , Proteínas del Citoesqueleto , Citometría de Flujo , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Fosfoproteínas/metabolismo , Bazo
9.
Cell Rep ; 38(11): 110507, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35294888

RESUMEN

Macrophage adhesion and stretching have been shown to induce interleukin (IL)-1ß production, but the mechanism of this mechanotransduction remains unclear. Here we specify the molecular link between mechanical tension on tissue-resident macrophages and activation of the NLRP3 inflammasome, which governs IL-1ß production. NLRP3 activation enhances antimicrobial defense, but excessive NLRP3 activity causes inflammatory tissue damage in conditions such as pulmonary fibrosis and acute respiratory distress syndrome. We find that the actin-bundling protein L-plastin (LPL) significantly enhances NLRP3 assembly. Specifically, LPL enables apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC) oligomerization during NLRP3 assembly by stabilizing ASC interactions with the kinase Pyk2, a component of cell-surface adhesive structures called podosomes. Upon treatment with exogenous NLRP3 activators, lung-resident alveolar macrophages (AMs) lacking LPL exhibit reduced caspase-1 activity, IL-1ß cleavage, and gasdermin-D processing. LPL-/- mice display resistance to bleomycin-induced lung injury and fibrosis. These findings identify the LPL-Pyk2-ASC pathway as a target for modulation in NLRP3-mediated inflammatory conditions.


Asunto(s)
Inflamasomas , Fibrosis Pulmonar , Animales , Bleomicina , Caspasa 1/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Mecanotransducción Celular , Glicoproteínas de Membrana , Ratones , Proteínas de Microfilamentos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fibrosis Pulmonar/inducido químicamente
10.
Sci Adv ; 5(8): eaax3387, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31453341

RESUMEN

Pneumonia poses profound health threats to preterm infants. Alveolar macrophages (AMs) eliminate inhaled pathogens while maintaining surfactant homeostasis. As AM development only occurs perinatally, therapies that accelerate AM maturation in preterms may improve outcomes. We tested therapeutic rescue of AM development in mice lacking the actin-bundling protein L-plastin (LPL), which exhibit impaired AM development and increased susceptibility to pneumococcal lung infection. Airway administration of recombinant granulocyte-macrophage colony-stimulating factor (GM-CSF) to LPL-/- neonates augmented AM production. Airway administration distinguishes the delivery route from prior human infant trials. Adult LPL-/- animals that received neonatal GM-CSF were protected from experimental pneumococcal challenge. No detrimental effects on surfactant metabolism or alveolarization were observed. Airway recombinant GM-CSF administration thus shows therapeutic promise to accelerate neonatal pulmonary immunity, protecting against bacterial pneumonia.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Macrófagos Alveolares/citología , Proteínas de Microfilamentos/genética , Neumonía Bacteriana/prevención & control , Administración por Inhalación , Animales , Animales Recién Nacidos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología
11.
Vaccine ; 35(34): 4389-4395, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28687405

RESUMEN

BACKGROUND: The epidemiology of nasopharyngeal (NP) pneumococcal carriage varies with geography and has changed in response to pneumococcal conjugate vaccine (PCV): a low prevalence (3% or less of colonizing isolates) of colonization by vaccine-type (VT) pneumococcal serotypes after PCV introduction has been reported. The primary goal of this study was to determine the VT serotype prevalence of NP pneumococcal colonization of children residing in the St. Louis, MO, USA metropolitan area following introduction of the 13-valent PCV in 2010. The secondary goal of this study was to identify characteristics associated with NP pneumococcal carriage of any serotype. METHODS: Between July 2013 and April 2016, we enrolled 397 healthy children, aged 0-17years, who required sedation for procedures or minor surgeries at St. Louis Children's Hospital. NP swabs were collected after sedation or anesthesia and cultured for pneumococcus. Vaccine records were obtained from primary care providers or from state immunization databases. Parents/guardians completed a questionnaire to provide demographics, past medical history and household characteristics. RESULTS: Of the 88 pneumococcal isolates recovered from 84 colonized subjects (21.2% of all enrolled subjects; 95% CI 17.2-25.2%), 16 were VT. Eleven isolates were serotype 19F (12.5%), four (4.5%) were 6A and one (1.1%) was 19A. Prevalence of VT among colonizing isolates was thus 18.2% (CI 10.1-26.1%) in our cohort, despite complete PCV vaccination in 87% of colonized children. Factors associated with pneumococcal colonization by any serotype included younger age and daycare attendance. CONCLUSION: Children in St. Louis exhibit a higher prevalence of VT serotypes among pneumococcal carriage isolates than has been reported in other areas in the US, demonstrating the necessity of ongoing surveillance of local epidemiology and providing evidence that serotype 19F can remain prevalent in a pediatric population despite high vaccine uptake.


Asunto(s)
Portador Sano/epidemiología , Infecciones Neumocócicas/epidemiología , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/aislamiento & purificación , Adolescente , Portador Sano/microbiología , Niño , Preescolar , Femenino , Vacuna Neumocócica Conjugada Heptavalente/administración & dosificación , Humanos , Lactante , Recién Nacido , Masculino , Pruebas de Sensibilidad Microbiana , Missouri/epidemiología , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/clasificación , Prevalencia , Estudios Seroepidemiológicos , Serogrupo , Streptococcus pneumoniae/clasificación , Streptococcus pneumoniae/inmunología , Vacunación , Vacunas Conjugadas/administración & dosificación
12.
Mol Immunol ; 78: 79-88, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27614263

RESUMEN

Elucidating the molecular regulation of macrophage migration is essential for understanding the pathophysiology of multiple human diseases, including host responses to infection and autoimmune disorders. Macrophage migration is supported by dynamic rearrangements of the actin cytoskeleton, with formation of actin-based structures such as podosomes and lamellipodia. Here we provide novel insights into the function of the actin-bundling protein l-plastin (LPL) in primary macrophages. We found that podosome stability is disrupted in primary resident peritoneal macrophages from LPL-/- mice. Live-cell imaging of F-actin using resident peritoneal macrophages from LifeACT-RFP+ mice demonstrated that loss of LPL led to decreased longevity of podosomes, without reducing the number of podosomes initiated. Additionally, macrophages from LPL-/- mice failed to elongate in response to chemotactic stimulation. These deficiencies in podosome stabilization and in macrophage elongation correlated with impaired macrophage transmigration in culture and decreased monocyte migration into murine peritoneum. Thus, we have identified a role for LPL in stabilizing long-lived podosomes and in enabling macrophage motility.


Asunto(s)
Movimiento Celular/fisiología , Macrófagos Peritoneales/metabolismo , Fosfoproteínas/metabolismo , Podosomas/metabolismo , Animales , Proteínas del Citoesqueleto , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Microscopía Confocal
13.
Int J Infect Dis ; 39: 50-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26327122

RESUMEN

Nasopharyngeal (NP) pneumococcal carriage predisposes children to pneumococcal infections. Defining the proportion of pneumococcal isolates that are antibiotic-resistant enables the appropriate choice of empiric therapies. The antibiogram of NP carriage isolates derived from a pediatric population following the introduction of the 13-valent pneumococcal conjugate vaccine was defined in this study.


Asunto(s)
Farmacorresistencia Bacteriana , Nasofaringe/microbiología , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/aislamiento & purificación , Adolescente , Antibacterianos/farmacología , Portador Sano/microbiología , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Infecciones Neumocócicas/epidemiología , Vacunas Neumococicas , Prevalencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA