Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Mol Immunol ; 18(1): 194-205, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31659245

RESUMEN

The adaptive immune response relies on specific apoptotic programs to maintain homeostasis. Conventional effector T cell (Tcon) expansion is constrained by both forkhead box P3 (FOXP3)+-regulatory T cells (Tregs) and restimulation-induced cell death (RICD), a propriocidal apoptosis pathway triggered by repeated stimulation through the T-cell receptor (TCR). Constitutive FOXP3 expression protects Tregs from RICD by suppressing SLAM-associated protein (SAP), a key adaptor protein that amplifies TCR signaling strength. The role of transient FOXP3 induction in activated human CD4 and CD8 Tcons remains unresolved, but its expression is inversely correlated with acquired RICD sensitivity. Here, we describe a novel role for FOXP3 in protecting human Tcons from premature RICD during expansion. Unlike FOXP3-mediated protection from RICD in Tregs, FOXP3 protects Tcons through a distinct mechanism requiring de novo transcription that does not require SAP suppression. Transcriptome profiling and functional analyses of expanding Tcons revealed that FOXP3 enhances expression of the SLAM family receptor CD48, which in turn sustains basal autophagy and suppresses pro-apoptotic p53 signaling. Both CD48 and FOXP3 expression reduced p53 accumulation upon TCR restimulation. Furthermore, silencing FOXP3 expression or blocking CD48 decreased the mitochondrial membrane potential in expanding Tcons with a concomitant reduction in basal autophagy. Our findings suggest that FOXP3 governs a distinct transcriptional program in early-stage effector Tcons that maintains RICD resistance via CD48-dependent protective autophagy and p53 suppression.


Asunto(s)
Antígeno CD48/metabolismo , Muerte Celular , Factores de Transcripción Forkhead/metabolismo , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/metabolismo , Linfocitos T Reguladores/inmunología , Apoptosis , Autofagia , Antígeno CD48/genética , Factores de Transcripción Forkhead/genética , Humanos , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Front Immunol ; 9: 2078, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30283440

RESUMEN

The caspase recruitment domain family member 11 (CARD11 or CARMA1)-B cell CLL/lymphoma 10 (BCL10)-MALT1 paracaspase (MALT1) [CBM] signalosome complex serves as a molecular bridge between cell surface antigen receptor signaling and the activation of the NF-κB, JNK, and mTORC1 signaling axes. This positions the CBM complex as a critical regulator of lymphocyte activation, proliferation, survival, and metabolism. Inborn errors in each of the CBM components have now been linked to a diverse group of human primary immunodeficiency diseases termed "CBM-opathies." Clinical manifestations range from severe combined immunodeficiency to selective B cell lymphocytosis, atopic disease, and specific humoral defects. This surprisingly broad spectrum of phenotypes underscores the importance of "tuning" CBM signaling to preserve immune homeostasis. Here, we review the distinct clinical and immunological phenotypes associated with human CBM complex mutations and introduce new avenues for targeted therapeutic intervention.


Asunto(s)
Proteína 10 de la LLC-Linfoma de Células B/inmunología , Proteínas Adaptadoras de Señalización CARD/inmunología , Guanilato Ciclasa/inmunología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Transducción de Señal/inmunología , Proteína 10 de la LLC-Linfoma de Células B/genética , Proteína 10 de la LLC-Linfoma de Células B/metabolismo , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Homeostasis/genética , Homeostasis/inmunología , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/genética , Proteína 1 de la Translocación del Linfoma del Tejido Linfático Asociado a Mucosas/metabolismo , Mutación/inmunología , Unión Proteica , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/metabolismo , Transducción de Señal/genética
3.
Front Immunol ; 9: 198, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29472930

RESUMEN

B-cell expansion with NF-κB and T-cell anergy (BENTA) disease is a B-cell-specific lymphoproliferative disorder caused by germline gain-of-function mutations in CARD11. These mutations force the CARD11 scaffold into an open conformation capable of stimulating constitutive NF-κB activation in lymphocytes, without requiring antigen receptor engagement. Many BENTA patients also suffer from recurrent infections, with 7 out of 16 patients exhibiting chronic, low-grade Epstein-Barr virus (EBV) viremia. In this mini-review, we discuss EBV infection in the pathogenesis and clinical management of BENTA disease, and speculate on mechanisms that could explain inadequate control of viral infection in BENTA patients.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Trastornos Linfoproliferativos/virología , FN-kappa B/inmunología , Linfocitos B/patología , Proteínas Adaptadoras de Señalización CARD/genética , Proliferación Celular , Anergia Clonal , Guanilato Ciclasa/genética , Herpesvirus Humano 4 , Humanos , Activación de Linfocitos , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/fisiopatología , Mutación , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Linfocitos T/inmunología , Viremia
4.
Front Immunol ; 9: 2944, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619304

RESUMEN

CARD11 is a lymphocyte-specific scaffold molecule required for proper activation of B- and T-cells in response to antigen. Germline gain-of-function (GOF) mutations in the CARD11 gene cause a unique B cell lymphoproliferative disorder known as B cell Expansion with NF-κB and T cell Anergy (BENTA). In contrast, patients carrying loss-of-function (LOF), dominant negative (DN) CARD11 mutations present with severe atopic disease. Interestingly, both GOF and DN CARD11 variants cause primary immunodeficiency, with recurrent bacterial and viral infections, likely resulting from impaired adaptive immune responses. This report describes a unique four-generation family harboring a novel heterozygous germline indel mutation in CARD11 (c.701-713delinsT), leading to one altered amino acid and a deletion of 4 others (p.His234_Lys238delinsLeu). Strikingly, affected members exhibit both moderate B cell lymphocytosis and atopic dermatitis/allergies. Ectopic expression of this CARD11 variant stimulated constitutive NF-κB activity in T cell lines, similar to other BENTA patient mutations. However, unlike other GOF mutants, this variant significantly impeded the ability of wild-type CARD11 to induce NF-κB activation following antigen receptor ligation. Patient lymphocytes display marked intrinsic defects in B cell differentiation and reduced T cell responsiveness in vitro. Collectively, these data imply that a single heterozygous CARD11 mutation can convey both GOF and DN signaling effects, manifesting in a blended BENTA phenotype with atopic features. Our findings further emphasize the importance of balanced CARD11 signaling for normal immune responses.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Mutación con Ganancia de Función , Mutación de Línea Germinal , Guanilato Ciclasa/genética , Síndromes de Inmunodeficiencia/genética , Trastornos Linfoproliferativos/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Proteínas Adaptadoras de Señalización CARD/metabolismo , Salud de la Familia , Femenino , Guanilato Ciclasa/metabolismo , Humanos , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Lactante , Trastornos Linfoproliferativos/patología , Masculino , FN-kappa B/metabolismo , Linaje , Linfocitos T/inmunología , Linfocitos T/metabolismo
6.
Front Immunol ; 8: 913, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28824638

RESUMEN

B cell Expansion with NF-κB and T cell Anergy (BENTA) disease is a novel B cell lymphoproliferative disorder caused by germline, gain-of-function mutations in the lymphocyte scaffolding protein CARD11, which drives constitutive NF-κB signaling. Despite dramatic polyclonal expansion of naive and immature B cells, BENTA patients also present with signs of primary immunodeficiency, including markedly reduced percentages of class-switched/memory B cells and poor humoral responses to certain vaccines. Using purified naive B cells from our BENTA patient cohort, here we show that BENTA B cells exhibit intrinsic defects in B cell differentiation. Despite a profound in vitro survival advantage relative to normal donor B cells, BENTA patient B cells were severely impaired in their ability to differentiate into short-lived IgDloCD38hi plasmablasts or CD138+ long-lived plasma cells in response to various stimuli. These defects corresponded with diminished IgG antibody production and correlated with poor induction of specific genes required for plasma cell commitment. These findings provide important mechanistic clues that help explain both B cell lymphocytosis and humoral immunodeficiency in BENTA disease.

7.
Nat Genet ; 49(8): 1192-1201, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28628108

RESUMEN

Few monogenic causes for severe manifestations of common allergic diseases have been identified. Through next-generation sequencing on a cohort of patients with severe atopic dermatitis with and without comorbid infections, we found eight individuals, from four families, with novel heterozygous mutations in CARD11, which encodes a scaffolding protein involved in lymphocyte receptor signaling. Disease improved over time in most patients. Transfection of mutant CARD11 expression constructs into T cell lines demonstrated both loss-of-function and dominant-interfering activity upon antigen receptor-induced activation of nuclear factor-κB and mammalian target of rapamycin complex 1 (mTORC1). Patient T cells had similar defects, as well as low production of the cytokine interferon-γ (IFN-γ). The mTORC1 and IFN-γ production defects were partially rescued by supplementation with glutamine, which requires CARD11 for import into T cells. Our findings indicate that a single hypomorphic mutation in CARD11 can cause potentially correctable cellular defects that lead to atopic dermatitis.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Dermatitis Atópica/genética , Mutación de Línea Germinal , Guanilato Ciclasa/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Estudios de Cohortes , Análisis Mutacional de ADN , Dermatitis Atópica/inmunología , Femenino , Genes Dominantes , Glutamina/metabolismo , Humanos , Células Jurkat , Activación de Linfocitos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Antígenos de Histocompatibilidad Menor/metabolismo , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Linaje , Linfocitos T/inmunología , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
8.
J Immunol ; 198(1): 147-155, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27852741

RESUMEN

Restimulation-induced cell death (RICD) regulates immune responses by restraining effector T cell expansion and limiting nonspecific damage to the host. RICD is triggered by re-engagement of the TCR on a cycling effector T cell, resulting in apoptosis. It remains unclear how RICD sensitivity is calibrated in T cells derived from different individuals or subsets. In this study we show that aerobic glycolysis strongly correlates with RICD sensitivity in human CD8+ effector T cells. Reducing glycolytic activity or glucose availability rendered effector T cells significantly less sensitive to RICD. We found that active glycolysis specifically facilitates the induction of proapoptotic Fas ligand upon TCR restimulation, accounting for enhanced RICD sensitivity in highly glycolytic T cells. Collectively, these data indicate that RICD susceptibility is linked to metabolic reprogramming, and that switching back to metabolic quiescence may help shield T cells from RICD as they transition into the memory pool.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD8-positivos/metabolismo , Glucólisis/inmunología , Western Blotting , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Vaccine ; 34(34): 4050-5, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27291087

RESUMEN

Epstein-Barr virus (EBV) is the primary cause of infectious mononucleosis and has been strongly implicated in the etiology of multiple epithelial and lymphoid cancers, such as nasopharyngeal carcinoma, gastric carcinoma, Hodgkin lymphoma, Burkitt lymphoma, non-Hodgkin lymphoma and post-transplant lymphoproliferative disorder. There is currently no licensed prophylactic vaccine for EBV. Most efforts to develop prophylactic vaccines have focused on EBV gp350, which binds to CD21/CD35 to gain entry into B cells, and is a major target of serum neutralizing antibody in EBV seropositive humans. However, a recombinant monomeric gp350 protein failed to prevent EBV infection in a phase II clinical trial. Thus, alternative or additional target antigens may be necessary for a successful prophylactic vaccine. EBV gH/gL and gB proteins coordinately mediate EBV fusion and entry into B cells and epithelial cells, strongly suggesting that vaccination with these proteins might elicit antibodies that will prevent EBV infection. We produced recombinant trimeric and monomeric EBV gH/gL heterodimeric proteins and a trimeric EBV gB protein, in addition to tetrameric and monomeric gp350(1-470) proteins, in Chinese hamster ovary cells. We demonstrated that vaccination of rabbits with trimeric and monomeric gH/gL, trimeric gB, and tetrameric gp350(1-470) induced serum EBV-neutralizing titers, using cultured human B cells, that were >100-fold, 20-fold, 18-fold, and 4-fold higher, respectively, than monomeric gp350(1-470). These data strongly suggest a role for testing EBV gH/gL and EBV gB in a future prophylactic vaccine to prevent EBV infection of B cells, as well as epithelial cells.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Virus de Epstein-Barr/prevención & control , Glicoproteínas de Membrana/inmunología , Chaperonas Moleculares/inmunología , Proteínas Virales/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Linfocitos B/inmunología , Linfocitos B/virología , Células CHO , Células Cultivadas , Cricetulus , Humanos , Masculino , Pruebas de Neutralización , Conejos , Proteínas Recombinantes
10.
J Immunol ; 196(11): 4614-21, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27183619

RESUMEN

Priming of mice with intact, heat-killed cells of Gram-negative Neisseria meningitidis, capsular serogroup C (MenC) or Gram-positive group B Streptococcus, capsular type III (GBS-III) bacteria resulted in augmented serum polysaccharide (PS)-specific IgG titers following booster immunization. Induction of memory required CD4(+) T cells during primary immunization. We determined whether PS-specific memory for IgG production was contained within the B cell and/or T cell populations, and whether augmented IgG responses following booster immunization were also dependent on CD4(+) T cells. Adoptive transfer of purified B cells from MenC- or GBS-III-primed, but not naive mice resulted in augmented PS-specific IgG responses following booster immunization. Similar responses were observed when cotransferred CD4(+) T cells were from primed or naive mice. Similarly, primary immunization with unencapsulated MenC or GBS-III, to potentially prime CD4(+) T cells, failed to enhance PS-specific IgG responses following booster immunization with their encapsulated isogenic partners. Furthermore, in contrast to GBS-III, depletion of CD4(+) T cells during secondary immunization with MenC or another Gram-negative bacteria, Acinetobacter baumannii, did not inhibit augmented PS-specific IgG booster responses of mice primed with heat-killed cells. Also, in contrast with GBS-III, booster immunization of MenC-primed mice with isolated MenC-PS, a TI Ag, or a conjugate of MenC-PS and tetanus toxoid elicited an augmented PS-specific IgG response similar to booster immunization with intact MenC. These data demonstrate that memory for augmented PS-specific IgG booster responses to Gram-negative and Gram-positive bacteria is contained solely within the B cell compartment, with a differential requirement for CD4(+) T cells for augmented IgG responses following booster immunization.


Asunto(s)
Inmunoglobulina G/inmunología , Neisseria meningitidis/inmunología , Polisacáridos/inmunología , Streptococcus agalactiae/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neisseria meningitidis/clasificación , Streptococcus agalactiae/clasificación
11.
Curr Opin Allergy Clin Immunol ; 15(6): 533-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26406182

RESUMEN

PURPOSE OF REVIEW: To present recent advances in the discovery and characterization of new immunodeficiency disorders linked to gain-of-function (GOF) mutations in immune signaling molecules. (Figure is included in full-text article.) RECENT FINDINGS: In the past 2 years, extensive cellular and molecular studies have illuminated the root causes of pathogenesis for several new monogenic primary immunodeficiency disorders (PIDs) linked to GOF mutations in signaling molecules. Here we discuss on two disorders (BENTA and APDS/PASLI) featuring shared clinical presentation (e.g. lymphoproliferation, selective antibody deficiencies, recurrent sinopulmonary infections). These findings highlight an emerging theme: both loss-of-function and gain-of-function mutations in key molecules can disrupt finely tuned immunoreceptor signaling modalities, resulting in the dysregulation of lymphocyte differentiation and impaired adaptive immunity. SUMMARY: Continued research on the molecular pathogenesis of PIDs defined by hyperactive signaling molecules will better distinguish these and related disorders, and pinpoint tailored therapeutic interventions for 'retuning' the immune response in these patients.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Síndromes de Inmunodeficiencia/inmunología , Linfocitos/inmunología , Mutación/genética , FN-kappa B/genética , Inmunidad Adaptativa/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Humanos , Síndromes de Inmunodeficiencia/genética , Terapia Molecular Dirigida , Enfermedades de Inmunodeficiencia Primaria , Transducción de Señal/genética , Transducción de Señal/inmunología
12.
Infect Immun ; 81(9): 3426-33, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23817619

RESUMEN

We previously demonstrated that intact, inactivated Streptococcus pneumoniae (unencapsulated strain R36A) inhibits IgG responses to a number of coimmunized soluble antigens (Ags). In this study, we investigated the mechanism of this inhibition and whether other extracellular bacteria exhibited similar effects. No inhibition was observed if R36A was given 24 h before or after immunization with soluble chicken ovalbumin (cOVA), indicating that R36A acts transiently during the initiation of the immune response. Using transgenic cOVA-specific CD4(+) T cells, we observed that R36A had no significant effect on T-cell activation (24 h) or generation of regulatory T cells (day 7) and only a modest effect on T-cell proliferation (48 to 96 h) in response to cOVA. However, R36A mediated a significant reduction in the formation of Ag-specific splenic germinal center T follicular helper (GC Tfh) and GC B cells and antibody-secreting cells in the spleen and bone marrow in response to cOVA or cOVA conjugated to 4-hydroxy-3-nitrophenylacetyl hapten (NP-cOVA). Of note, the inhibitory effect of intact R36A on the IgG anti-cOVA response could be reproduced using R36A-derived cell walls. In contrast to R36A, neither inactivated, unencapsulated, intact Neisseria meningitidis nor Streptococcus agalactiae inhibited the OVA-specific IgG response. These results suggest a novel immunosuppressive property within the cell wall of Streptococcus pneumoniae.


Asunto(s)
Centro Germinal/inmunología , Inmunosupresores/inmunología , Células Plasmáticas/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/metabolismo , Células Productoras de Anticuerpos/inmunología , Células Productoras de Anticuerpos/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Pared Celular/inmunología , Pared Celular/metabolismo , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Centro Germinal/metabolismo , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunosupresores/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Células Plasmáticas/metabolismo , Infecciones Neumocócicas/metabolismo , Infecciones Neumocócicas/microbiología , Bazo/inmunología , Bazo/metabolismo , Streptococcus pneumoniae/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
13.
J Immunol ; 189(2): 575-86, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22706079

RESUMEN

Murine IgG responses specific for the capsular polysaccharide (pneumococcal capsular polysaccharide serotype 14; PPS14) of Streptococcus pneumoniae type 14 (Pn14), induced in response to intact Pn14 or a PPS14-protein conjugate, are both dependent on CD4(+) T cell help but appear to use marginal zone versus follicular B cells, respectively. In this study, we identify an idiotype (44.1-Id) that dominates the PPS14-specific IgG, but not IgM, responses to intact Pn14, isolated PPS14, and Group B Streptococcus (strain COH1-11) expressing capsular polysaccharide structurally identical to PPS14. The 44.1-Id, however, is not expressed in the repertoire of natural PPS14-specific Abs. In distinct contrast, PPS14-specific IgG responses to a soluble PPS14-protein conjugate exhibit minimal usage of the 44.1-Id, although significant 44.1-Id expression is elicited in response to conjugate attached to particles. The 44.1-Id elicited in response to intact Pn14 was expressed in similar proportions among all four IgG subclasses during both the primary and secondary responses. The 44.1-Id usage was linked to the Igh(a), but not Igh(b), allotype and was associated with induction of relatively high total PPS14-specific IgG responses. In contrast to PPS14-protein conjugate, avidity maturation of the 44.1-Id-dominant PPS14-specific IgG responses was limited, even during the highly boosted T cell-dependent PPS14-specific secondary responses to COH1-11. These results indicate that different antigenic forms of the same capsular polysaccharide can recruit distinct B cell clones expressing characteristic idiotypes under genetic control and suggest that the 44.1-Id is derived from marginal zone B cells.


Asunto(s)
Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/inmunología , Idiotipos de Inmunoglobulinas/biosíntesis , Vacunas Neumococicas/inmunología , Vacunas Conjugadas/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/metabolismo , Cápsulas Bacterianas/administración & dosificación , Sitios de Unión de Anticuerpos , Femenino , Idiotipos de Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Vacunas Neumococicas/administración & dosificación , Streptococcus pneumoniae/inmunología , Vacunas Conjugadas/administración & dosificación
14.
J Immunol ; 188(11): 5238-46, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22523389

RESUMEN

We previously reported distinct differences in the murine in vivo Ig polysaccharide (PS)-specific responses to intact Streptococcus pneumoniae compared with responses to Neisseria meningitidis and that in each case, the bacterial subcapsular domain markedly influences the Ig response to the associated PS. In light of potentially unique contributions of biochemically distinct capsular PS and/or their characteristic attachments to the underlying bacterium, it remains unresolved whether different bacterial subcapsular domains can exert differential effects on PS-specific Ig responses to distinct bacterial pathogens. In this report, we used a mutant strain of group B Streptococcus (Streptococcus agalactiae) type III (GBS-III) that expresses desialylated capsular polysaccharide of GBS-III, biochemically identical to capsular pneumococcal polysaccharide type 14 (PPS14) of Streptococcus pneumoniae (intact inactivated Streptococcus pneumoniae, capsular type 14, Pn14), directly to compare the in vivo PPS14-specific IgG responses to two distinct gram-positive bacteria. Although both GBS-III and Pn14 elicited relatively rapid primary PPS14-specific IgG responses dependent on CD4(+) T cells, B7-dependent costimulation, and CD40-CD40L interactions, only GBS-III induced a highly boosted ICOS-dependent PPS14-specific IgG response after secondary immunization. Of note, priming with Pn14 and boosting with GBS-III, although not isolated PPS14, elicited a similar boosted PPS14-specific IgG response that was dependent on CD4(+) T cells during secondary immunization, indicating that Pn14 primes for memory but, unlike GBS-III, fails to elicit it. The inability of Pn14 to elicit a boosted PPS14-specific IgG response was overcome by coimmunization with unencapsulated GBS-III. Collectively, these data establish that structurally identical capsular PS expressed by two distinct gram-positive extracellular bacteria can indeed elicit distinct PS-specific IgG responses in vivo.


Asunto(s)
Cápsulas Bacterianas/biosíntesis , Cápsulas Bacterianas/química , Inmunoglobulina G/biosíntesis , Polisacáridos Bacterianos/biosíntesis , Polisacáridos Bacterianos/química , Streptococcus agalactiae/inmunología , Streptococcus pneumoniae/inmunología , Animales , Cápsulas Bacterianas/inmunología , Femenino , Inmunización Secundaria , Ratones , Ratones Endogámicos BALB C , Estructura Terciaria de Proteína , Especificidad de la Especie , Vacunas Estreptocócicas/administración & dosificación , Vacunas Estreptocócicas/inmunología , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/inmunología
15.
J Immunol ; 188(2): 569-77, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22156342

RESUMEN

In vivo anti-polysaccharide Ig responses to isolated polysaccharide (PS) are T cell independent, rapid, and fail to generate memory. However, little is known regarding PS-specific Ig responses to intact gram-positive and gram-negative extracellular bacteria. We previously demonstrated that intact heat-killed Streptococcus pneumoniae, a gram-positive bacterium, elicited a rapid primary pneumococcal capsular PS (PPS) response in mice that was dependent on CD4(+) T cells, B7-dependent costimulation, and CD40-CD40L interactions. However, this response was ICOS independent and failed to generate a boosted PPS-specific secondary IgG response. In the current study, we analyzed the murine meningococcal type C PS (MCPS)-specific Ig response to i.p.-injected intact, heat-killed Neisseria meningitidis, serogroup C (MenC), a gram-negative bacterium. In contrast to S. pneumoniae, the IgG anti-MCPS response to MenC exhibited delayed primary kinetics and was highly boosted after secondary immunization, whereas the IgG anti-MCPS response to isolated MCPS was rapid, without secondary boosting, and consisted of only IgG1 and IgG3, as opposed to all four IgG isotypes in response to intact MenC. The secondary, but not primary, IgG anti-MCPS response to MenC was dependent on CD4(+) T cells, CD40L, CD28, and ICOS. The primary and secondary IgG anti-MCPS responses were lower in TLR4-defective (C3H/HeJ) but not TLR2(-/-) or MyD88(-/-) mice, but secondary boosting was still observed. Of interest, coimmunization of S. pneumoniae and MenC resulted in a boosted secondary IgG anti-PPS response to S. pneumoniae. Our data demonstrate that the nature of the in vivo anti-PS response is markedly influenced by the composition and/or architecture of the bacterial subcapsular domain.


Asunto(s)
Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/inmunología , Neisseria meningitidis Serogrupo C/inmunología , Polisacáridos Bacterianos/inmunología , Animales , Antígenos Bacterianos/administración & dosificación , Cápsulas Bacterianas/administración & dosificación , Células Cultivadas , Femenino , Inmunización Secundaria , Inmunoglobulina G/biosíntesis , Lipopolisacáridos/administración & dosificación , Lipopolisacáridos/inmunología , Vacunas Meningococicas/administración & dosificación , Vacunas Meningococicas/inmunología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Ratones Transgénicos , Polisacáridos Bacterianos/administración & dosificación , Estructura Terciaria de Proteína , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...