Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Cell ; 82(16): 3077-3088.e6, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35738262

RESUMO

Glycolysis facilitates the rapid recall response of CD8+ memory T (Tm) cells. However, it remains unclear whether Tm cells uptake exogenous glucose or mobilize endogenous sugar to fuel glycolysis. Here, we show that intracellular glycogen rather than extracellular glucose acts as the major carbon source for the early recall response. Following antigenic stimulation, Tm cells exhibit high glycogen phosphorylase (brain form, PYGB) activity, leading to glycogenolysis and release of glucose-6-phosphate (G6P). Elevated G6P mainly flows to glycolysis but is also partially channeled to the pentose phosphate pathway, which maintains the antioxidant capacity necessary for later recall stages. Mechanistically, TCR signaling directly induces phosphorylation of PYGB by LCK-ZAP70. Functionally, the glycogenolysis-fueled early recall response of CD8+ Tm cells accelerates the clearance of OVA-Listeria monocytogenes in an infected mouse model. Thus, we uncover a specific dependency on glycogen for the initial activation of memory T cells, which may have therapeutic implications for adaptive immunity.


Assuntos
Glicogenólise , Animais , Linfócitos T CD8-Positivos , Glucose/metabolismo , Glicogênio/metabolismo , Células T de Memória , Camundongos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo
2.
J Infect Dis ; 227(10): 1203-1213, 2023 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-36408618

RESUMO

BACKGROUND: Although modified vaccinia Ankara-Bavarian Nordic (MVA-BN) vaccination is approved for smallpox and monkeypox prevention, immunological persistence and booster effects remain undescribed. METHODS: Participants naive to smallpox vaccination were randomized to 1 dose MVA-BN (1×MVA, n = 181), 2 doses MVA-BN (2×MVA, n = 183), or placebo (n = 181). Participants with previous smallpox vaccination received 1 MVA-BN booster (HSPX, n = 200). Subsets of the formerly naive groups (approximately 75 each) received an MVA-BN booster 2 years later. RESULTS: Neutralizing antibody (nAb) geometric mean titers (GMTs) increased from 1.1 (baseline, both naive groups) to 7.2 and 7.5 (week 4, 1×MVA and 2×MVA, respectively), and further to 45.6 (week 6, 2×MVA after second vaccination). In HSPX, nAb GMT rapidly increased from 21.6 (baseline) to 175.1 (week 2). At 2 years, GMTs for 1×MVA, 2×MVA, and HSPX were 1.1, 1.3, and 10.3, respectively. After boosting in the previously naive groups, nAb GMTs increased rapidly in 2 weeks to 80.7 (1×MVA) and 125.3 (2×MVA), higher than after primary vaccination and comparable to boosted HSPX subjects. Six months after boosting, GMTs were 25.6 (1×MVA) and 49.3 (2×MVA). No safety concerns were identified. CONCLUSIONS: Anamnestic responses to boosting without sustained high nAb titers support presence of durable immunological memory following primary MVA-BN immunization. Clinical Trials Registration. NCT00316524 and NCT00686582.


Assuntos
Vacina Antivariólica , Varíola , Vacínia , Humanos , Varíola/prevenção & controle , Anticorpos Antivirais , Vaccinia virus , Vacinação , Anticorpos Neutralizantes
3.
J Allergy Clin Immunol ; 145(5): 1309-1321, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32386655

RESUMO

Impaired vaccine responses in older individuals are associated with alterations in both the quantity and quality of the T-cell compartment with age. As reviewed herein, the T-cell response to vaccination requires a fine balance between the generation of inflammatory effector T cells versus follicular helper T (TFH) cells that mediate high-affinity antibody production in tandem with the induction of long-lived memory cells for effective recall immunity. During aging, we find that this balance is tipped where T cells favor short-lived effector but not memory or TFH responses. Consistently, vaccine-induced antibodies commonly display a lower protective capacity. Mechanistically, multiple, potentially targetable, changes in T cells have been identified that contribute to these age-related defects, including posttranscription regulation, T-cell receptor signaling, and metabolic function. Although research into the induction of tissue-specific immunity by vaccines and with age is still limited, current mechanistic insights provide a framework for improved design of age-specific vaccination strategies that require further evaluation in a clinical setting.


Assuntos
Envelhecimento/imunologia , Vacinação , Animais , Linfócitos B/imunologia , Humanos , Vacinas
4.
Adv Exp Med Biol ; 1254: 55-62, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32323269

RESUMO

Memory B cells are a key cellular component of the protective humoral responses to infectious pathogens. Most of our knowledge of memory B-cell responses comes from studies using mono-epitopic model antigens that elicit systemic humoral responses dominated by canonical B-cell antigen receptors. This approach successfully dissected the systemic responses of memory B cells and greatly advanced our understanding of memory B-cell formation, maintenance, and reactivation to re-invading antigens in the secondary lymphoid organs. However, the canonical memory B-cell responses fail to fully recapitulate the heterogeneity of the protective memory responses. Indeed, accumulating studies using "natural" antigens and live pathogens have uncovered new aspects of memory B-cell responses, which are achieved by memory B cells with different phenotypes, tissue residence, and responsiveness to antigen stimulation. Such non-canonical memory B-cell responses are frequently observed in local sites where live pathogens initially infect and replicate. Importantly, the local memory B-cell responses often serve as the first line of defense against re-infecting pathogens, thereby playing an essential role in controlling the pathogens. Here, we provide a comprehensive overview of the systemic and local memory B-cell responses in the humoral protective immunity against pathogens.


Assuntos
Linfócitos B , Memória Imunológica , Animais , Antígenos , Humanos , Imunidade Humoral , Receptores de Antígenos de Linfócitos B
5.
Helicobacter ; 24(5): e12652, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31414552

RESUMO

BACKGROUND: Tissue-resident memory T cells accelerate the clearance of pathogens during recall response. However, whether CD4+ TRM cells themselves can provide gastric immunity is unclear. MATERIALS AND METHODS: We established a parabiosis model between the enhanced green fluorescent protein and wild-type mice that the circulation system was shared, and the wild-type partner was vaccinated with H pylori vaccine composed of CCF and silk fibroin in gastric subserous layer to induce gastric EGFP+ CD4+ TRM cells. Antigen-specific EGFP+ CD4+ T cells and proliferous TRM cells were analyzed by flow cytometry. The colonization of H pylori was detected by quantitative real-time PCR. EGFP+ CD4+ TRM cells and the inflammation of the stomach were observed by histology. RESULTS: A parabiosis animal model was employed to identify the cells that introduced by vaccination in GSL. Antigen-specific EGFP+ CD4+ T cells could be detected at day 7 post-vaccination. Thirty days later, EGFP+ CD4+ TRM cells were established with a phenotype of CD69+ CD103- . Of note, we found that when circulating lymphocytes were depleted by FTY720 administration, these TRM cells could proliferate in situ and differentiate into effector Th1 cells after H pylori challenge. A decrease in H pylori colonization was observed in the vaccinated mice but not unvaccinated mice. Further, we found that although FTY720 was administrated, mounted pro-inflammatory myeloid cells still emerged in the stomach of the vaccinated mice, which might contribute to the reduction of H pylori colonization. CONCLUSIONS: Our study reveals that H pylori vaccine-induced CD4+ TRM cells can proliferate and differentiate in situ to enhance gastric local immunity during recall response.


Assuntos
Vacinas Bacterianas/imunologia , Linfócitos T CD4-Positivos/imunologia , Mucosa Gástrica/imunologia , Infecções por Helicobacter/prevenção & controle , Helicobacter pylori/imunologia , Memória Imunológica , Animais , Vacinas Bacterianas/administração & dosagem , Proliferação de Células , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Camundongos Endogâmicos C57BL , Subpopulações de Linfócitos T/imunologia
6.
Int Immunol ; 28(6): 267-82, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26714588

RESUMO

Memory CD4(+) T cells promote protective humoral immunity; however, how memory T cells acquire this activity remains unclear. This study demonstrates that CD4(+) T cells develop into antigen-specific memory T cells that can promote the terminal differentiation of memory B cells far more effectively than their naive T-cell counterparts. Memory T cell development requires the transcription factor B-cell lymphoma 6 (Bcl6), which is known to direct T-follicular helper (Tfh) cell differentiation. However, unlike Tfh cells, memory T cell development did not require germinal center B cells. Curiously, memory T cells that develop in the absence of cognate B cells cannot promote memory B-cell recall responses and this defect was accompanied by down-regulation of genes associated with homeostasis and activation and up-regulation of genes inhibitory for T-cell responses. Although memory T cells display phenotypic and genetic signatures distinct from Tfh cells, both had in common the expression of a group of genes associated with metabolic pathways. This gene expression profile was not shared to any great extent with naive T cells and was not influenced by the absence of cognate B cells during memory T cell development. These results suggest that memory T cell development is programmed by stepwise expression of gatekeeper genes through serial interactions with different types of antigen-presenting cells, first licensing the memory lineage pathway and subsequently facilitating the functional development of memory T cells. Finally, we identified Gdpd3 as a candidate genetic marker for memory T cells.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Diester Fosfórico Hidrolases/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Antígenos CD4/metabolismo , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Imunocompetência , Memória Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Transcriptoma
7.
Front Immunol ; 14: 1228532, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37868996

RESUMO

Introduction: Immunotherapies have shown great promise, but are not effective for all tumors types and are effective in less than 3% of patients with pancreatic ductal adenocarcinomas (PDAC). To make an immune treatment that is effective for more cancer patients and those with PDAC specifically, we genetically engineered Salmonella to deliver exogenous antigens directly into the cytoplasm of tumor cells. We hypothesized that intracellular delivery of an exogenous immunization antigen would activate antigen-specific CD8 T cells and reduce tumors in immunized mice. Methods: To test this hypothesis, we administered intracellular delivering (ID) Salmonella that deliver ovalbumin as a model antigen into tumor-bearing, ovalbumin-vaccinated mice. ID Salmonella delivers antigens by autonomously lysing in cells after the induction of cell invasion. Results: We showed that the delivered ovalbumin disperses throughout the cytoplasm of cells in culture and in tumors. This delivery into the cytoplasm is essential for antigen cross-presentation. We showed that co-culture of ovalbumin-recipient cancer cells with ovalbumin-specific CD8 T cells triggered a cytotoxic T cell response. After the adoptive transfer of OT-I CD8 T cells, intracellular delivery of ovalbumin reduced tumor growth and eliminated tumors. This effect was dependent on the presence of the ovalbumin-specific T cells. Following vaccination with the exogenous antigen in mice, intracellular delivery of the antigen cleared 43% of established KPC pancreatic tumors, increased survival, and prevented tumor re-implantation. Discussion: This response in the immunosuppressive KPC model demonstrates the potential to treat tumors that do not respond to checkpoint inhibitors, and the response to re-challenge indicates that new immunity was established against intrinsic tumor antigens. In the clinic, ID Salmonella could be used to deliver a protein antigen from a childhood immunization to refocus pre-existing T cell immunity against tumors. As an off-the-shelf immunotherapy, this bacterial system has the potential to be effective in a broad range of cancer patients.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , Criança , Ovalbumina , Camundongos Endogâmicos C57BL , Antígenos de Neoplasias/metabolismo , Vacinação , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Salmonella/genética
8.
Front Immunol ; 13: 842870, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35418979

RESUMO

Intestinal parasitic nematodes affect a quarter of the world's population, typically eliciting prominent effector Th2-driven host immune responses. As not all infected hosts develop protection against reinfection, our current understanding of nematode-induced memory Th2 responses remains limited. Here, we investigated the activation of memory Th2 cells and the mechanisms driving early recall responses to the enteric nematode Heligmosomoides polygyrus in mice. We show that nematode-cured mice harbor memory Th2 cells in lymphoid and non-lymphoid organs with distinct transcriptional profiles, expressing recirculation markers like CCR7 and CD62-L in the mesenteric lymph nodes (mLN), and costimulatory markers like Ox40, as well as tissue homing and activation markers like CCR2, CD69 and CD40L in the gut and peritoneal cavity (PEC). While memory Th2 cells persist systemically in both lymphoid and non-lymphoid tissues following cure of infection, peritoneal memory Th2 cells in particular displayed an initial prominent expansion and strong parasite-specific Th2 responses during early recall responses to a challenge nematode infection. This effect was paralleled by a significant influx of dendritic cells (DC) and eosinophils, both also appearing exclusively in the peritoneal cavity of reinfected mice. In addition, we show that within the peritoneal membrane lined by peritoneal mesothelial cells (PeM), the gene expression levels of cell adhesion markers VCAM-1 and ICAM-1 decrease significantly in response to a secondary infection. Overall, our findings indicate that the host peritoneal cavity in particular harbors prominent memory Th2 cells and appears to respond directly to H. polygyrus by an early recall response via differential regulation of cell adhesion markers, marking the peritoneal cavity an important site for host immune responses to an enteric pathogen.


Assuntos
Nematospiroides dubius , Infecções por Strongylida , Animais , Linfonodos , Camundongos , Cavidade Peritoneal , Células Th2
9.
Neurol Int ; 14(4): 943-951, 2022 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-36412697

RESUMO

Anti-CD20 therapies decrease the humoral response to SARS-CoV-2 immunization. We aimed to determine the extent of the humoral response to SARS-CoV-2 antigens in correlation with peripheral B-cell dynamics among patients with central nervous system inflammatory disorders treated with anti-CD20 medications. We retrospectively included patients receiving anti-CD20 therapy after antigen contact who were divided into responders (>7 binding antibody units (BAU)/mL) and non-responders (<7 BAU/mL). In participants with first antigen contact prior to therapy, we investigated the recall response elicited once under treatment. We included 80 patients (responders n = 34, non-responders n = 37, recall cohort n = 9). The B-cell counts among responders were significantly higher compared to non-responders (mean 1012 cells/µL ± SD 105 vs. mean 17 cells/µL ± SD 47; p < 0.001). Despite very low B-cell counts (mean 9 cells/µL ± SD 20), humoral response was preserved among the recall cohort (mean 1653 BAU/mL ± SD 2250.1) and did not differ significantly from responders (mean 735 BAU/mL ± SD 1529.9; p = 0.14). Our data suggest that peripheral B cells are required to generate antibodies to neo-antigens but not for a recall response during anti-CD20 therapy. Evaluation of B-cell counts and pre-existing SARS-CoV-2 antibodies might serve as biomarkers for estimating the immune competence to mount a humoral response to SARS-CoV-2 antigens.

10.
Front Immunol ; 11: 624199, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33815352

RESUMO

Resident memory CD8+ T (TRM) cells are a lymphocyte lineage distinct from circulating memory CD8+ T cells. TRM lodge within peripheral tissues and secondary lymphoid organs where they provide rapid, local protection from pathogens and control tumor growth. However, dysregulation of CD8+ TRM formation and/or activation may contribute to the pathogenesis of autoimmune diseases. Intrinsic mechanisms, including transcriptional networks and inhibitory checkpoint receptors control TRM differentiation and response. Additionally, extrinsic stimuli such as cytokines, cognate antigen, fatty acids, and damage signals regulate TRM formation, maintenance, and expansion. In this review, we will summarize knowledge of CD8+ TRM generation and highlight mechanisms that regulate the persistence and responses of heterogeneous TRM populations in different tissues and distinct microenvironments.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Memória Imunológica , Neoplasias/imunologia , Transdução de Sinais/imunologia , Microambiente Tumoral/imunologia , Linfócitos T CD8-Positivos/patologia , Humanos , Neoplasias/patologia , Especificidade de Órgãos/imunologia
11.
Artigo em Inglês | MEDLINE | ID: mdl-32477964

RESUMO

Mounting experimental evidence hints to an import role for natural killer (NK) cells in adaptive immune responses to pathogens. NK cells with adaptive features are heterogeneous and belong to different subsets according to their phenotype as well as the nature of their adaptive recall reactions. Three types of adaptive NK cell responses have been described: (i) NK cells with long-lived memory of multiple different haptens and viral antigens were described in murine liver tissue with a possible human counterpart; (ii) infection of human and mouse cytomegalovirus is associated with an expansion of NKG2C+ and Ly49H+ NK cells, respectively, that selectively recognize CMV-encoded peptides thereby facilitating recall responses; (iii) cytokine-stimulated NK cells respond to different stimuli with enhanced production of IFN-γ after re-stimulation. These exciting findings not only support the idea of NK cells with adaptive features, but define a novel field of harnessing memory NK cell subsets for therapeutic strategies.


Assuntos
Citomegalovirus , Muromegalovirus , Animais , Antígenos Virais , Células Matadoras Naturais , Camundongos , Fenótipo
12.
Elife ; 82019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31225793

RESUMO

In mice, memory B (Bmem) cells can be divided into two subpopulations: CD80hi Bmem cells, which preferentially differentiate into plasma cells; and CD80lo Bmem cells, which become germinal center (GC) B cells during a recall response. We demonstrate that these distinct responses can be B-cell-intrinsic and essentially independent of B-cell receptor (BCR) isotypes. Furthermore, we find that the development of CD80hi Bmem cells in the primary immune response requires follicular helper T cells, a relatively strong CD40 signal and a high-affinity BCR on B cells, whereas the development of CD80lo Bmem cells does not. Quantitative differences in CD40 stimulation were enough to recapitulate the distinct B cell fate decisions in an in vitro culture system. The quantity of CD40 signaling appears to be translated into NF-κB activation, followed by BATF upregulation that promotes Bmem cell differentiation from GC B cells.


Assuntos
Linfócitos B/imunologia , Antígenos CD40/imunologia , Memória Imunológica/genética , Receptores de Antígenos de Linfócitos B/genética , Animais , Antígeno B7-1/genética , Antígenos CD40/genética , Diferenciação Celular/genética , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Centro Germinativo/imunologia , Isotipos de Imunoglobulinas , Memória Imunológica/imunologia , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , Plasmócitos/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/imunologia
13.
Front Immunol ; 10: 2262, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31608063

RESUMO

Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency characterized by recurrent infections, micro thrombocytopenia, eczema, and a high incidence of autoimmunity and malignancy. A defect in the T cell compartment is thought to be a major cause of immunodeficiency in patients with WAS; However, whether the antigen specific T memory cell is altered has not been extensively studied. Here, we examined the expansion/contraction kinetics of CD8+ memory T cells and their maintenance in WASp-/- mice. The results showed that WAS protein (WASp) is not required for differentiation of CD8+ effector T cells; however, CD8+ T cells from WASp-/- mice were hyperactive, resulting in increased cytokine production. The number of CD8+ T memory cells decreased as mice aged, and CD8+ T cell recall responses and protective immunity were impaired. WASp-deficient CD8+ T cells in bone marrow chimeric mice underwent clonal expansion, but the resulting effector cells failed to survive and differentiate into CD8+ memory T cells. Taken together, these findings indicate that WASp plays an intrinsic role in differentiation of CD8+ memory T cells.


Assuntos
Apoptose/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Proteína da Síndrome de Wiskott-Aldrich/imunologia , Animais , Apoptose/genética , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proliferação de Células/genética , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Memória Imunológica/genética , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Camundongos Knockout , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
14.
mSphere ; 4(1)2019 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-30814319

RESUMO

Microneme (MIC) proteins play important roles in the recognition, adhesion, and invasion of host cells by Toxoplasma gondii Previous studies have shown that MIC proteins are highly immunogenic in the mouse and recognized by human serum antibodies. Here we report that T. gondii antigens MIC1, MIC3, MIC4, and MIC6 were capable of inducing memory responses leading to production of gamma interferon (IFN-γ) by T cells from T. gondii-infected mice. Production of IFN-γ was demonstrated using enzyme-linked immunosorbent spot (ELISPOT) assay and also intracellular cytokine staining. All four MIC antigens displayed very high sensitivity (100%) and specificity (86 to 100%) for detecting chronic infection. Interestingly, IFN-γ was produced by both CD4+ and CD8+ T cells in BALB/c mice but primarily by CD4+ T cells in C57BL/6 mice. Phenotypic characterization of IFN-γ-producing CD4+ and CD8+ T cells in BALB/c mice and CD4+ T cells in C57BL/6 mice revealed effector memory T cells (CD44hi CD62Llo) as the predominant cells that contributed to IFN-γ production in response to MIC antigens. Effector memory responses were seen in mice of different major histocompatibility complex class II (MHC-II) haplotypes, suggesting that MIC antigens contain epitopes that are broadly recognized.IMPORTANCE Current diagnosis of toxoplasmosis relies almost exclusively on antibody detection, and while detection of IgG provides a useful estimate of prior infection, it does not alone indicate immune status. In contrast, detection of IFN-γ responses to T. gondii antigens has been used to monitor immune responsiveness in HIV-infected patients, thus providing valuable predictions about the potential for disease reactivation. However, specific T. gondii antigens that can be used in assays to detect cellular immunity remain largely undefined. In this study, we examined the diagnostic potential of microneme antigens of T. gondii using IFN-γ detection assays. Our findings demonstrate that MIC antigens (MIC1, MIC3, MIC4, and MIC6) elicit IFN-γ responses from memory T cells in chronically infected mice. Monitoring IFN-γ production by T cells stimulated with MIC antigens provided high sensitivity and specificity for detection of T. gondii infection in mice. Taken together, these studies suggest that microneme antigens might be useful as an adjunct to serological testing to monitor immune status during infection.


Assuntos
Memória Imunológica , Proteínas de Protozoários/imunologia , Linfócitos T/imunologia , Toxoplasmose/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Moléculas de Adesão Celular/imunologia , Doença Crônica , Feminino , Imunidade Celular , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Organismos Livres de Patógenos Específicos , Toxoplasma
15.
Front Immunol ; 7: 165, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27242779

RESUMO

The current influenza vaccine provides narrow protection against the strains included in the vaccine, and needs to be reformulated every few years in response to the constantly evolving new strains. Novel approaches are directed toward developing vaccines that provide broader protection by targeting B and T cell epitopes that are conserved between different strains of the virus. In this paper, we focus on developing mathematical models to explore the CD8 T cell responses to influenza, how they can be boosted, and the conditions under which they contribute to protection. Our models suggest that the interplay between spatial heterogeneity (with the virus infecting the respiratory tract and the immune response being generated in the secondary lymphoid organs) and T cell differentiation (with proliferation occurring in the lymphoid organs giving rise to a subpopulation of resident T cells in the respiratory tract) is the key to understand the dynamics of protection afforded by the CD8 T cell response to influenza. Our results suggest that the time lag for the generation of resident T cells in the respiratory tract and their rate of decay following infection are the key factors that limit the efficacy of CD8 T cell responses. The models predict that an increase in the level of central memory T cells leads to a gradual decrease in the viral load, and, in contrast, there is a sharper protection threshold for the relationship between the size of the population of resident T cells and protection. The models also suggest that repeated natural influenza infections cause the number of central memory CD8 T cells and the peak number of resident memory CD8 T cells to reach their plateaus, and while the former is maintained, the latter decays with time since the most recent infection.

16.
Cell Rep ; 17(3): 636-644, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27732841

RESUMO

Despite robust secondary T cell expansion primed by vaccination, the impact on primary immune responses to heterotypic antigens remains undefined. Here we show that secondary expansion of epitope-specific memory CD8+ T cells primed by prior infection with recombinant pathogens limits the primary expansion of naive CD8+ T cells with specificity to new heterologous antigens, dampening protective immunity against subsequent pathogen challenge. The degree of naive T cell repression directly paralleled the magnitude of the recall response. Suppressed primary T cell priming reflects competition for antigen accessibility, since clonal expansion was not inhibited if the primary and secondary epitopes were expressed on different dendritic cells. Interestingly, robust recall responses did not impact antigen-specific NK cells, suggesting that adaptive and innate lymphocyte responses possess different activation requirements or occur in distinct anatomical locations. These findings have important implications in pathogen vaccination strategies that depend on the targeting of multiple T cell epitopes.


Assuntos
Epitopos de Linfócito T/imunologia , Muromegalovirus/fisiologia , Linfócitos T/imunologia , Vacinação , Animais , Células Apresentadoras de Antígenos/metabolismo , Antígenos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Células Clonais , Listeria/fisiologia , Camundongos , Recombinação Genética/genética
17.
Thromb Haemost ; 114(6): 1127-35, 2015 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-26245154

RESUMO

Development of inhibitory antibodies against factor VIII (FVIII) is a severe complication of replacement therapy in haemophilia A. Patients with inhibitors are treated with high FVIII doses in the context of immune tolerance therapy (ITT). Data from haemophilia A mouse model suggest that high FVIII concentrations prevent the formation of antibody secreting cells (ASCs) from memory B cells (MBCs) by inducing apoptosis. Fc gamma receptor 2B (CD32) is an important regulator of B cell function, mediating inhibitory signals after cross-linking with the B cell receptor. Here, the role of CD32 in the regulation of FVIII-specific MBCs was investigated using F8-/- and F8-/-CD32-/- knockout mice and monoclonal antibodies (mAbs). The initial immune response was similar between F8-/- and F8-/-CD32-/- mice, including concentration of anti-FVIII antibodies and number of FVIII-specific ASCs in spleen and bone marrow. In contrast, formation of ASCs from MBCs upon rhFVIII re-stimulation in vitro was abolished in F8-/-CD32-/- mice, whereas FVIII/anti-FVIII immune complexes significantly enhanced ASC formation in F8-/- mice. Inhibition of CD32 by mAbs or F(ab)2 fragments prevented ASC formation in a dose-dependent manner. Transfer of B cell-depleted splenocytes using CD45R (B220) depletion from CD32-competent mice did not restore ASC formation in F8-/-CD32-/- cells confirming that CD32 is required on B cells. We conclude that CD32 is a crucial regulator of FVIII-specific B cells and is required for the differentiation of MBCs into ASCs. Inhibition of CD32 could potentially improve the efficacy of FVIII in the context of ITT.


Assuntos
Subpopulações de Linfócitos B/imunologia , Fator VIII/imunologia , Memória Imunológica/imunologia , Ativação Linfocitária/imunologia , Receptores de IgG/fisiologia , Transferência Adotiva , Animais , Anticorpos Heterófilos/biossíntese , Anticorpos Heterófilos/imunologia , Anticorpos Monoclonais , Anticorpos Neutralizantes/biossíntese , Anticorpos Neutralizantes/imunologia , Complexo Antígeno-Anticorpo/análise , Apoptose , Medula Óssea/imunologia , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Fator VIII/genética , Fator VIII/farmacologia , Fator VIII/uso terapêutico , Hemofilia A/imunologia , Antígenos Comuns de Leucócito/imunologia , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Knockout , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/deficiência , Receptores de IgG/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Baço/imunologia
18.
Pathog Dis ; 70(2): 141-52, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24453125

RESUMO

Global gene expression profile changes were monitored in human peripheral blood mononuclear cells (PBMCs) after challenge with the live vaccine strain (LVS) of Francisella tularensis. Because these PBMCs were from individuals previously immunized with LVS, stimulating these cells with LVS should activate memory responses. The Ingenuity Pathway Analysis tool identified pathways, functions, and networks associated with this in vitro recall response, including novel pathways triggered by the memory response. Dendritic cell (DC) maturation was the most significant among the more than 25 relevant pathways discovered. Interleukin 15, granulocyte-macrophage colony-stimulating factor, and triggering receptor expressed on myeloid cells 1 signaling pathways were also significant. Pathway analysis indicated that Class 1 antigen presentation may not be optimal with LVS vaccination. The top three biological functions were antigen presentation, cell-mediated and humoral immune responses. Network analysis revealed that the top network associated with these functions had IFNγ and TNFα in central interactive positions. Our results suggest that DC maturation is a key factor in the recall responses and that more effective antigen processing and presentation is needed for cytotoxic T lymphocyte responses. Taken together, these considerations are critical for future tularemia vaccine development studies.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Leucócitos Mononucleares/imunologia , Adolescente , Adulto , Diferenciação Celular , Células Cultivadas , Células Dendríticas/imunologia , Humanos , Memória Imunológica , Masculino , Pessoa de Meia-Idade , Adulto Jovem
19.
Braz. j. med. biol. res ; 46(2): 148-153, 01/fev. 2013. graf
Artigo em Inglês | LILACS | ID: lil-668770

RESUMO

Serogroup B Neisseria meningitidis (MenB) is a major cause of invasive disease in early childhood worldwide. The only MenB vaccine available in Brazil was produced in Cuba and has shown unsatisfactory efficacy when used to immunize millions of children in Brazil. In the present study, we compared the specific functional antibody responses evoked by the Cuban MenB vaccine with a standard vaccine against diphtheria (DTP: diphtheria, tetanus, pertussis) after primary immunization and boosting of mice. The peak of bactericidal and opsonic antibody titers to MenB and of neutralizing antibodies to diphtheria toxoid (DT) was reached after triple immunization with the MenB vaccine or DTP vaccine, respectively. However, 4 months after immunization, protective DT antibody levels were present in all DTP-vaccinated mice but in only 20% of the mice immunized against MenB. After 6 months of primary immunization, about 70% of animals still had protective neutralizing DT antibodies, but none had significant bactericidal antibodies to MenB. The booster doses of DTP or MenB vaccines produced a significant antibody recall response, suggesting that both vaccines were able to generate and maintain memory B cells during the period studied (6 months post-triple immunization). Therefore, due to the short duration of serological memory induced by the MenB vaccine (VA-MENGOC-BC® vaccine), its use should be restricted to outbreaks of meningococcal disease.


Assuntos
Animais , Feminino , Camundongos , Anticorpos Antibacterianos/imunologia , Toxoide Diftérico/imunologia , Vacina contra Difteria, Tétano e Coqueluche/imunologia , Infecções Meningocócicas/imunologia , Neisseria meningitidis/imunologia , Formação de Anticorpos , Antígenos de Bactérias/imunologia , Memória Imunológica , Infecções Meningocócicas/prevenção & controle , Vacinas Meningocócicas/imunologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa