Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Vaccines (Basel) ; 10(5)2022 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-35632507

RESUMO

Many bacterial infections are major health problems worldwide, and treatment of many of these infectious diseases is becoming increasingly difficult due to the development of antibiotic resistance, which is a major threat. Prophylactic vaccines against these bacterial pathogens are urgently needed. This is also true for bacterial infections that are still neglected, even though they affect a large part of the world's population, especially under poor hygienic conditions. One example is typhus, a life-threatening disease also known as "war plague" caused by Rickettsia prowazekii, which could potentially come back in a war situation such as the one in Ukraine. However, vaccination against bacterial infections is a challenge. In general, bacteria are much more complex organisms than viruses and as such are more difficult targets. Unlike comparatively simple viruses, bacteria possess a variety of antigens whose immunogenic potential is often unknown, and it is unclear which antigen can elicit a protective and long-lasting immune response. Several vaccines against extracellular bacteria have been developed in the past and are still used successfully today, e.g., vaccines against tetanus, pertussis, and diphtheria. However, while induction of antibody production is usually sufficient for protection against extracellular bacteria, vaccination against intracellular bacteria is much more difficult because effective defense against these pathogens requires T cell-mediated responses, particularly the activation of cytotoxic CD8+ T cells. These responses are usually not efficiently elicited by immunization with non-living whole cell antigens or subunit vaccines, so that other antigen delivery strategies are required. This review provides an overview of existing antibacterial vaccines and novel approaches to vaccination with a focus on immunization against intracellular bacteria.

3.
Vaccines (Basel) ; 9(8)2021 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-34452021

RESUMO

Rickettsioses are febrile, potentially lethal infectious diseases that are a serious health threat, especially in poor income countries. The causative agents are small obligate intracellular bacteria, rickettsiae. Rickettsial infections are emerging worldwide with increasing incidence and geographic distribution. Nonetheless, these infections are clearly underdiagnosed because methods of diagnosis are still limited and often not available. Another problem is that the bacteria respond to only a few antibiotics, so delayed or wrong antibiotic treatment often leads to a more severe outcome of the disease. In addition to that, the development of antibiotic resistance is a serious threat because alternative antibiotics are missing. For these reasons, prophylactic vaccines against rickettsiae are urgently needed. In the past years, knowledge about protective immunity against rickettsiae and immunogenic determinants has been increasing and provides a basis for vaccine development against these bacterial pathogens. This review provides an overview of experimental vaccination approaches against rickettsial infections and perspectives on vaccination strategies.

4.
PLoS One ; 16(6): e0253084, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34111210

RESUMO

Rickettsioses are neglected and emerging potentially fatal febrile diseases that are caused by obligate intracellular bacteria, rickettsiae. Rickettsia (R.) typhi and R. prowazekii constitute the typhus group (TG) of rickettsiae and are the causative agents of endemic and epidemic typhus, respectively. We recently generated a monoclonal antibody (BNI52) against R. typhi. Characterization of BNI52 revealed that it specifically recognizes TG rickettsiae but not the members of the spotted fever group (SFG) rickettsiae. We further show that BNI52 binds to protein fragments of ±30 kDa that are exposed on the bacterial surface and also present in the periplasmic space. These protein fragments apparently derive from the cytosolic GroEL protein of R. typhi and are also recognized by antibodies in the sera from patients and infected mice. Furthermore, BNI52 opsonizes the bacteria for the uptake by antigen presenting cells (APC), indicating a contribution of GroEL-specific antibodies to protective immunity. Finally, it is interesting that the GroEL protein belongs to 32 proteins that are differentially downregulated by R. typhi after passage through immunodeficient BALB/c CB17 SCID mice. This could be a hint that the rickettsia GroEL protein may have immunomodulatory properties as shown for the homologous protein from several other bacteria, too. Overall, the results of this study provide evidence that GroEL represents an immunodominant antigen of TG rickettsiae that is recognized by the humoral immune response against these pathogens and that may be interesting as a vaccine candidate. Apart from that, the BNI52 antibody represents a new tool for specific detection of TG rickettsiae in various diagnostic and experimental setups.


Assuntos
Anticorpos Monoclonais/metabolismo , Chaperonina 60/imunologia , Infecções por Rickettsia/sangue , Rickettsia typhi/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/metabolismo , Anticorpos Monoclonais/sangue , Antígenos de Bactérias/imunologia , Linhagem Celular , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Camundongos SCID , Periplasma/metabolismo , Infecções por Rickettsia/imunologia , Infecções por Rickettsia/microbiologia , Xenopus laevis
5.
PLoS Negl Trop Dis ; 14(10): e0008704, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33091016

RESUMO

Over the last decades, rickettsioses are emerging worldwide. These diseases are caused by intracellular bacteria. Although rickettsioses can be treated with antibiotics, a vaccine against rickettsiae is highly desired for several reasons. Rickettsioses are highly prevalent, especially in poor countries, and there are indications of the development of antibiotic resistance. In addition, some rickettsiae can persist and cause recurrent disease. The development of a vaccine requires the understanding of the immune mechanisms that are involved in protection as well as in immunopathology. Knowledge about these immune responses is accumulating, and efforts have been undertaken to identify antigenic components of rickettsiae that may be useful as a vaccine. This review provides an overview on current knowledge of adaptive immunity against rickettsiae, which is essential for defense, rickettsial antigens that have been identified so far, and on vaccination strategies that have been used in animal models of rickettsial infections.


Assuntos
Vacinas Bacterianas/imunologia , Infecções por Rickettsia/prevenção & controle , Rickettsiaceae/imunologia , Imunidade Adaptativa , Doenças Transmissíveis Emergentes/prevenção & controle , Humanos , Infecções por Rickettsia/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-29358295

RESUMO

Scrub typhus is a potentially lethal infection caused by the obligate intracellular bacterium Orientia tsutsugamushi Reports on the emergence of doxycycline-resistant strains highlight the urgent need to develop novel antiinfectives against scrub typhus. Corallopyronin A (CorA) is a novel α-pyrone compound synthesized by the myxobacterium Corallococcus coralloides that was characterized as a noncompetitive inhibitor of the switch region of the bacterial RNA polymerase (RNAP). We investigated the antimicrobial action of CorA against the human-pathogenic Karp strain of O. tsutsugamushiin vitro and in vivo The MIC of CorA against O. tsutsugamushi was remarkably low (0.0078 µg/ml), 16-fold lower than that against Rickettsia typhi In the lethal intraperitoneal O. tsutsugamushi mouse infection model, a minimum daily dose of 100 µg CorA protected 100% of infected mice. Two days of treatment were sufficient to confer protection. In contrast to BALB/c mice, SCID mice succumbed to the infection despite treatment with CorA or tetracycline, suggesting that antimicrobial treatment required synergistic action of the adaptive immune response. Similar to tetracycline, CorA did not prevent latent infection of O. tsutsugamushiin vivo However, latency was not caused by acquisition of antimicrobial resistance, since O. tsutsugamushi reisolated from latently infected BALB/c mice remained fully susceptible to CorA. No mutations were found in the CorA-binding regions of the ß and ß' RNAP subunit genes rpoB and rpoC Inhibition of the RNAP switch region of O. tsutsugamushi by CorA is therefore a novel and highly potent target for antimicrobial therapy for scrub typhus.


Assuntos
Antibacterianos/uso terapêutico , RNA Polimerases Dirigidas por DNA/antagonistas & inibidores , Lactonas/uso terapêutico , Orientia tsutsugamushi/patogenicidade , Tifo por Ácaros/tratamento farmacológico , Animais , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Myxococcales/efeitos dos fármacos , Myxococcales/patogenicidade , Orientia tsutsugamushi/efeitos dos fármacos , Rickettsia typhi/efeitos dos fármacos , Rickettsia typhi/patogenicidade , Tifo por Ácaros/microbiologia
7.
Med Microbiol Immunol ; 206(6): 403-417, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28770333

RESUMO

Rickettsiae are small intracellular bacteria that can cause life-threatening febrile diseases. Rickettsioses occur worldwide with increasing incidence. Therefore, a vaccine is highly desired. A prerequisite for the development of a vaccine is the knowledge of the immune response against these bacteria, in particular protective immunity. In recent years murine models of rickettsial infections have been established, and the study of immune response against rickettsiae in mice provided many new insights into protective and pathological immune reactions. This review summarizes the current knowledge about immune mechanisms in protection and pathology in rickettsial infections.


Assuntos
Imunidade Adaptativa , Modelos Animais de Doenças , Imunidade Inata , Infecções por Rickettsia/imunologia , Rickettsia/imunologia , Animais , Camundongos
8.
Oncotarget ; 8(22): 35542-35557, 2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28415767

RESUMO

Regulatory T cells (Tregs) are potential immunotherapeutic candidates to induce transplantation tolerance. However, stability of Tregs still remains contentious and may potentially restrict their clinical use. Recent work suggested that epigenetic imprinting of Foxp3 and other Treg-specific signature genes is crucial for stabilization of immunosuppressive properties of Foxp3+ Tregs, and that these events are initiated already during early stages of thymic Treg development. However, the mechanisms governing this process remain largely unknown. Here we demonstrate that thymic antigen-presenting cells (APCs), including thymic dendritic cells (t-DCs) and medullary thymic epithelial cells (mTECs), can induce a more pronounced demethylation of Foxp3 and other Treg-specific epigenetic signature genes in developing Tregs when compared to splenic DCs (sp-DCs). Transcriptomic profiling of APCs revealed differential expression of secreted factors and costimulatory molecules, however neither addition of conditioned media nor interference with costimulatory signals affected Foxp3 induction by thymic APCs in vitro. Importantly, when tested in vivo both mTEC- and t-DC-generated alloantigen-specific Tregs displayed significantly higher efficacy in prolonging skin allograft acceptance when compared to Tregs generated by sp-DCs. Our results draw attention to unique properties of thymic APCs in initiating commitment towards stable and functional Tregs, a finding that could be highly beneficial in clinical immunotherapy.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Epigênese Genética , Impressão Genômica , Isoantígenos/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Biomarcadores , Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Metilação de DNA , Feminino , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais , Transplante de Pele , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timócitos/imunologia , Timócitos/metabolismo , Timo/imunologia , Timo/metabolismo
9.
Infect Immun ; 85(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28289147

RESUMO

Rickettsia typhi is the causative agent of endemic typhus, a disease with increasing incidence worldwide that can be fatal. Because of its obligate intracellular life style, genetic manipulation of the pathogen is difficult. Nonetheless, in recent years, genetic manipulation tools have been successfully applied to rickettsiae. We describe here for the first time the transformation of R. typhi with the pRAM18dRGA plasmid that originally derives from Rickettsia amblyommatis and encodes the expression of GFPuv (green fluorescent protein with maximal fluorescence when excited by UV light). Transformed R. typhi (R. typhiGFPuv) bacteria are viable, replicate with kinetics similar to those of wild-type R. typhi in cell culture, and stably maintain the plasmid and GFPuv expression under antibiotic treatment in vitro and in vivo during infection of mice. CB17 SCID mice infected with R. typhiGFPuv succumb to the infection with kinetics similar to those for animals infected with wild-type R. typhi and develop comparable pathology and bacterial loads in the organs, demonstrating that the plasmid does not influence pathogenicity. In the spleen and liver of infected CB17 SCID mice, the bacteria are detectable by immunofluorescence microscopy in neutrophils and macrophages by histological staining. Finally, we show for the first time that transformed rickettsiae can be used for the detection of CD8+ T cell responses. GFP-specific restimulation of spleen cells from R. typhiGFPuv-infected BALB/c mice elicits gamma interferon (IFN-γ), tumor necrosis factor alpha (TNF-α), and interleukin 2 (IL-2) secretion by CD8+ T cells. Thus, R. typhiGFPuv bacteria are a novel, potent tool to study infection with the pathogen in vitro and in vivo and the immune response to these bacteria.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/imunologia , Rickettsia typhi/patogenicidade , Tifo Endêmico Transmitido por Pulgas/imunologia , Animais , Proteínas de Fluorescência Verde/genética , Fígado/microbiologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Neutrófilos/microbiologia , Plasmídeos , Rickettsia typhi/genética , Baço/microbiologia , Transformação Bacteriana , Tifo Endêmico Transmitido por Pulgas/microbiologia
10.
PLoS Negl Trop Dis ; 11(2): e0005404, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28222146

RESUMO

Endemic typhus caused by Rickettsia (R.) typhi is an emerging febrile disease that can be fatal due to multiple organ pathology. Here we analyzed the requirements for protection against R. typhi by T cells in the CB17 SCID model of infection. BALB/c wild-type mice generate CD4+ TH1 and cytotoxic CD8+ T cells both of which are sporadically reactivated in persistent infection. Either adoptively transferred CD8+ or CD4+ T cells protected R. typhi-infected CB17 SCID mice from death and provided long-term control. CD8+ T cells lacking either IFNγ or Perforin were still protective, demonstrating that the cytotoxic function of CD8+ T cells is not essential for protection. Immune wild-type CD4+ T cells produced high amounts of IFNγ, induced the release of nitric oxide in R. typhi-infected macrophages and inhibited bacterial growth in vitro via IFNγ and TNFα. However, adoptive transfer of CD4+IFNγ-/- T cells still protected 30-90% of R. typhi-infected CB17 SCID mice. These cells acquired a TH17 phenotype, producing high amounts of IL-17A and IL-22 in addition to TNFα, and inhibited bacterial growth in vitro. Surprisingly, the neutralization of either TNFα or IL-17A in CD4+IFNγ-/- T cell recipient mice did not alter bacterial elimination by these cells in vivo, led to faster recovery and enhanced survival compared to isotype-treated animals. Thus, collectively these data show that although CD4+ TH1 cells are clearly efficient in protection against R. typhi, CD4+ TH17 cells are similarly protective if the harmful effects of combined production of TNFα and IL-17A can be inhibited.


Assuntos
Citocinas/metabolismo , Rickettsia typhi/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th1/imunologia , Células Th17/imunologia , Tifo Endêmico Transmitido por Pulgas/imunologia , Tifo Endêmico Transmitido por Pulgas/patologia , Transferência Adotiva , Animais , Modelos Animais de Doenças , Camundongos Endogâmicos BALB C , Camundongos SCID
11.
Med Microbiol Immunol ; 206(1): 41-51, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27696011

RESUMO

Rickettsioses are caused by intracellular bacteria of the family of Rickettsiaceae. Rickettsia (R.) typhi is the causative agent of endemic typhus. The disease occurs worldwide and is one of the most prevalent rickettsioses. Rickettsial diseases, however, are generally underdiagnosed which is mainly due to the lack of sensitive and specific methods. In addition, methods for quantitative detection of the bacteria for research purposes are rare. We established two qPCRs for the detection of R. typhi by amplification of the outer membrane protein B (ompB) and parvulin-type PPIase (prsA) genes. Both qPCRs are specific and exclusively recognize R. typhi but no other rickettsiae including the closest relative, R. prowazekii. The prsA-based qPCR revealed to be much more sensitive than the amplification of ompB and provided highly reproducible results in the detection of R. typhi in organs of infected mice. Furthermore, as a nested PCR the prsA qPCR was applicable for the detection of R. typhi in human blood samples. Collectively, the prsA-based qPCR represents a reliable method for the quantitative detection of R. typhi for research purposes and is a promising candidate for differential diagnosis.


Assuntos
Técnicas de Diagnóstico Molecular/métodos , Reação em Cadeia da Polimerase em Tempo Real/métodos , Rickettsia typhi/isolamento & purificação , Tifo Endêmico Transmitido por Pulgas/diagnóstico , Animais , Proteínas da Membrana Bacteriana Externa/genética , Proteínas de Bactérias/genética , Sangue/microbiologia , Humanos , Lipoproteínas/genética , Proteínas de Membrana/genética , Camundongos SCID , Reprodutibilidade dos Testes , Rickettsia typhi/genética , Sensibilidade e Especificidade , Tifo Endêmico Transmitido por Pulgas/microbiologia
12.
PLoS Negl Trop Dis ; 10(11): e0005089, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27875529

RESUMO

Rickettsia typhi is an intracellular bacterium that causes endemic typhus, a febrile disease that can be fatal due to complications including pneumonia, hepatitis and meningoencephalitis, the latter being a regular outcome in T and B cell-deficient C57BL/6 RAG1-/- mice upon Rickettsia typhi infection. Here, we show that CD4+ TH1 cells that are generated in C57BL/6 mice upon R. typhi infection are as protective as cytotoxic CD8+ T cells. CD4+- as well as CD8+-deficient C57BL/6 survived the infection without showing symptoms of disease at any point in time. Moreover, adoptively transferred CD8+ and CD4+ immune T cells entered the CNS of C57BL/6 RAG1-/- mice with advanced infection and both eradicated the bacteria. However, immune CD4+ T cells protected only approximately 60% of the animals from death. They induced the expression of iNOS in infiltrating macrophages as well as in resident microglia in the CNS which can contribute to bacterial killing but also accelerate pathology. In vitro immune CD4+ T cells inhibited bacterial growth in infected macrophages which was in part mediated by the release of IFNγ. Collectively, our data demonstrate that CD4+ T cells are as protective as CD8+ T cells against R. typhi, provided that CD4+ TH1 effector cells are present in time to support bactericidal activity of phagocytes via the release of IFNγ and other factors. With regard to vaccination against TG Rickettsiae, our findings suggest that the induction of CD4+ TH1 effector cells is sufficient for protection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Macrófagos/imunologia , Rickettsia typhi/imunologia , Tifo Endêmico Transmitido por Pulgas/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD8-Positivos/microbiologia , Feminino , Humanos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Rickettsia typhi/fisiologia , Células Th1/imunologia , Células Th1/microbiologia , Tifo Endêmico Transmitido por Pulgas/microbiologia
13.
PLoS Negl Trop Dis ; 10(8): e0004935, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27548618

RESUMO

Rickettsia (R.) typhi is the causative agent of endemic typhus, an emerging febrile disease that is associated with complications such as pneumonia, encephalitis and liver dysfunction. To elucidate how innate immune mechanisms contribute to defense and pathology we here analyzed R. typhi infection of CB17 SCID mice that are congenic to BALB/c mice but lack adaptive immunity. CB17 SCID mice succumbed to R. typhi infection within 21 days and showed high bacterial load in spleen, brain, lung, and liver. Most evident pathological changes in R. typhi-infected CB17 SCID mice were massive liver necrosis and splenomegaly due to the disproportionate accumulation of neutrophils and macrophages (MΦ). Both neutrophils and MΦ infiltrated the liver and harbored R. typhi. Both cell populations expressed iNOS and produced reactive oxygen species (ROS) and, thus, exhibited an inflammatory and bactericidal phenotype. Surprisingly, depletion of neutrophils completely prevented liver necrosis but neither altered bacterial load nor protected CB17 SCID mice from death. Furthermore, the absence of neutrophils had no impact on the overwhelming systemic inflammatory response in these mice. This response was predominantly driven by activated MΦ and NK cells both of which expressed IFNγ and is considered as the reason of death. Finally, we observed that iNOS expression by MΦ and neutrophils did not correlate with R. typhi uptake in vivo. Moreover, we demonstrate that MΦ hardly respond to R. typhi in vitro. These findings indicate that R. typhi enters MΦ and also neutrophils unrecognized and that activation of these cells is mediated by other mechanisms in the context of tissue damage in vivo.


Assuntos
Inflamação/imunologia , Inflamação/patologia , Células Matadoras Naturais/imunologia , Fígado/patologia , Macrófagos/imunologia , Neutrófilos/imunologia , Tifo Endêmico Transmitido por Pulgas/imunologia , Animais , Encéfalo/microbiologia , Citocinas/biossíntese , Modelos Animais de Doenças , Imunidade Inata , Inflamação/sangue , Inflamação/microbiologia , Interferon gama/biossíntese , Fígado/microbiologia , Pulmão/microbiologia , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Necrose , Óxido Nítrico Sintase Tipo II/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Rickettsia typhi/imunologia , Rickettsia typhi/patogenicidade , Baço/microbiologia , Baço/patologia , Tifo Endêmico Transmitido por Pulgas/microbiologia , Tifo Endêmico Transmitido por Pulgas/patologia
14.
Infect Immun ; 84(5): 1615-1632, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26975992

RESUMO

Rickettsioses are emerging febrile diseases caused by obligate intracellular bacteria belonging to the family Rickettsiaceae. Rickettsia typhi belongs to the typhus group (TG) of this family and is the causative agent of endemic typhus, a disease that can be fatal. In the present study, we analyzed the course of R. typhi infection in C57BL/6 RAG1(-/-) mice. Although these mice lack adaptive immunity, they developed only mild and temporary symptoms of disease and survived R. typhi infection for a long period of time. To our surprise, 3 to 4 months after infection, C57BL/6 RAG1(-/-) mice suddenly developed lethal neurological disorders. Analysis of these mice at the time of death revealed high bacterial loads, predominantly in the brain. This was accompanied by a massive expansion of microglia and by neuronal cell death. Furthermore, high numbers of infiltrating CD11b(+) macrophages were detectable in the brain. In contrast to the microglia, these cells harbored R. typhi and showed an inflammatory phenotype, as indicated by inducible nitric oxide synthase (iNOS) expression, which was not observed in the periphery. Having shown that R. typhi persists in immunocompromised mice, we finally asked whether the bacteria are also able to persist in resistant C57BL/6 and BALB/c wild-type mice. Indeed, R. typhi could be recultivated from lung, spleen, and brain tissues from both strains even up to 1 year after infection. This is the first report demonstrating persistence and reappearance of R. typhi, mainly restricted to the central nervous system in immunocompromised mice.


Assuntos
Infecções do Sistema Nervoso Central/microbiologia , Infecções do Sistema Nervoso Central/patologia , Inflamação/patologia , Rickettsia typhi/isolamento & purificação , Tifo Endêmico Transmitido por Pulgas/microbiologia , Tifo Endêmico Transmitido por Pulgas/patologia , Animais , Encéfalo/parasitologia , Encéfalo/patologia , Doença Crônica , Proteínas de Homeodomínio/genética , Pulmão/parasitologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/análise , Baço/parasitologia
15.
Immunobiology ; 220(8): 964-75, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25766204

RESUMO

CD83 is a transmembrane glycoprotein that is rapidly up-regulated on activated B cells. Although CD83 itself is incapable to transduce intracellular signaling, it acts as a negative regulator of B cell function. We have recently described that a single application of anti-CD83 antibody results in dramatically enhanced production of antigen-specific IgG1 but not other isotypes upon immunization of mice with the TI-2 model antigen (Ag) NIP-Ficoll. This effect was mediated by the binding of anti-CD83 to CD83 on the surface of B cells themselves. In the current study we show that administration of anti-CD83 enhances IgG1-production independent of IL-4. Application of anti-CD83 does not alter the proliferation and general expansion of NIP-specific B cells. In the presence of anti-CD83, immunized mice develop normal frequencies of plasmablasts in response to NIP-Ficoll of which an increased number produces IgG1. These cells localize in extrafollicular foci in the spleen of immunized mice and originate from the marginal zone B cell pool. Taken together, our results indicate that CD83 engagement in vivo does not generally enhance B cell activation but selectively promotes IgG1 class switch in marginal zone B cells in response to TI-2 Ag.


Assuntos
Antígenos CD/imunologia , Antígenos T-Independentes/imunologia , Linfócitos B/imunologia , Switching de Imunoglobulina/imunologia , Imunoglobulinas/imunologia , Glicoproteínas de Membrana/imunologia , Animais , Imunoglobulina G/metabolismo , Interleucina-4/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Superfície Celular/genética , Baço/imunologia , Regulação para Cima , Antígeno CD83
16.
J Neuroinflammation ; 11: 86, 2014 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-24886419

RESUMO

BACKGROUND: Increasing evidence suggests that inflammation associated with microglial cell activation in the substantia nigra (SN) of patients with Parkinson disease (PD) is not only a consequence of neuronal degeneration, but may actively sustain dopaminergic (DA) cell loss over time. We aimed to study whether the intracellular chaperone heat shock protein 60 (Hsp60) could serve as a signal of CNS injury for activation of microglial cells. METHODS: Hsp60 mRNA expression in the mesencephalon and the striatum of C57/BL6 mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and the Hsp60/TH mRNA ratios in the SN of PD patients and aged-matched subjects were measured. To further investigate a possible link between the neuronal Hsp60 response and PD-related cellular stress, Hsp60 immunoblot analysis and quantification in cell lysates from SH-SY5Y after treatment with 100 µM MPP+ (1-methyl-4-phenylpyridinium) at different time points (6, 12, 24 and 48 hours) compared to control cells were performed. Additional MTT and LDH assay were used. We next addressed the question as to whether Hsp60 influences the survival of TH+ neurons in mesencephalic neuron-glia cultures treated either with MPP+ (1 µM), hHsp60 (10 µg/ml) or a combination of both. Finally, we measured IL-1ß, IL-6, TNF-α and NO-release by ELISA in primary microglial cell cultures following treatment with different hHsp60 preparations. Control cultures were exposed to LPS. RESULTS: In the mesencephalon and striatum of mice treated with MPTP and also in the SN of PD patients, we found that Hsp60 mRNA was up-regulated. MPP+, the active metabolite of MPTP, also caused an increased expression and release of Hsp60 in the human dopaminergic cell line SH-SY5Y. Interestingly, in addition to being toxic to DA neurons in primary mesencephalic cultures, exogenous Hsp60 aggravated the effects of MPP+. Yet, although we demonstrated that Hsp60 specifically binds to microglial cells, it failed to stimulate the production of pro-inflammatory cytokines or NO by these cells. CONCLUSIONS: Overall, our data suggest that Hsp60 is likely to participate in DA cell death in PD but via a mechanism unrelated to cytokine release.


Assuntos
Chaperonina 60/metabolismo , Corpo Estriado/patologia , Neurônios Dopaminérgicos/metabolismo , Intoxicação por MPTP/patologia , Mesencéfalo/patologia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Chaperonina 60/genética , Modelos Animais de Doenças , Dopaminérgicos/farmacologia , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Óxido Nítrico/metabolismo , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
17.
Int Immunol ; 25(9): 507-20, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23728778

RESUMO

Transgenic over-expression of CD83 on B cells leads to a reduced response to BCR engagement but to an enhanced secretion of IL-10 upon LPS stimulation. In this study, we analyzed the differential influence of CD83 on the stimulation of different B cell subsets via the BCR or TLR4. Neither wild type nor CD83 transgenic (CD83tg) B cells produced any IL-10 in response to BCR stimulation. BCR engagement led to reduced activation of LYN, SYK and ERK1/2 resulting in reduced numbers of proliferating cells in all CD83tg B cell subsets. Moreover, CD83tg follicular (FO) but not marginal zone (MZ) or transitional (TN) B cells showed significantly enhanced cell death. In contrast, LPS stimulation led to normal frequencies of proliferating CD83tg FO, MZ and TN B cells although TLR4 engagement did not rescue FO B cells from apoptosis. Furthermore, LPS stimulation led to high IL-10 production derived from CD83tg MZ B cells that reacted to LPS stimulation with enhanced ERK1/2 activation. Finally, we show that CD83 co-localizes with the BCR complex as well as with the LPS receptor complex suggesting that CD83 interacts with components of both signaling complexes. Taken together, the results of this study show that CD83 already inhibits the initiation of BCR signaling leading to insufficient activation signals in all B cells and reduced survival especially of FO B cells. On the other hand, CD83 supports TLR4-mediated IL-10 release exclusively in MZ B cells. Thus, CD83 differentially modulates FO and MZ B cell responses.


Assuntos
Antígenos CD/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Baço/citologia , Baço/imunologia , Animais , Antígenos CD/imunologia , Imunoglobulinas/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Antígeno CD83
18.
Monoclon Antib Immunodiagn Immunother ; 32(2): 98-104, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23607344

RESUMO

CD83 is expressed on mature dendritic cells (DCs) and many immune cells, including B and T lymphocytes upon activation. CD83 is essential for the development of CD4 single positive T cells and involved in the regulation of peripheral T and B cell responses. Here we generated a new monoclonal antibody against human CD83, which we named ELBE-1. ELBE-1 recognizes a linear epitope and cannot only be applied for Western blot analysis and flow cytometry, but also detects soluble CD83 (sCD83) by ELISA. As with other anti-CD83 antibodies described thus far, ELBE-1 does not influence T cell activation. Nevertheless, it represents a useful new tool for the detection and functional characterization of CD83.


Assuntos
Anticorpos Monoclonais Murinos/química , Antígenos CD/imunologia , Imunoglobulinas/imunologia , Glicoproteínas de Membrana/imunologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Western Blotting , Células COS , Proliferação de Células , Chlorocebus aethiops , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Células Alimentadoras , Feminino , Citometria de Fluxo , Humanos , Hibridomas , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Linfócitos T/fisiologia , Antígeno CD83
19.
Sci Rep ; 3: 1393, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23462811

RESUMO

In mammalians, toll-like receptors (TLR) signal-transduction pathways induce the expression of a variety of immune-response genes, including inflammatory cytokines. It is therefore plausible to assume that TLRs are mediators in glial cells triggering the release of cytokines that ultimately kill DA neurons in the substantia nigra in Parkinson disease (PD). Accordingly, recent data indicate that TLR4 is up-regulated by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment in a mouse model of PD. Here, we wished to evaluate the role of TLR4 in the acute mouse MPTP model of PD: TLR4-deficient mice and wild-type littermates control mice were used for the acute administration way of MPTP or a corresponding volume of saline. We demonstrate that TLR4-deficient mice are less vulnerable to MPTP intoxication than wild-type mice and display a decreased number of Iba1+ and MHC II+ activated microglial cells after MPTP application, suggesting that the TLR4 pathway is involved in experimental PD.


Assuntos
Transtornos Parkinsonianos/metabolismo , Receptor 4 Toll-Like/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/efeitos adversos , Animais , Morte Celular/genética , Corpo Estriado/metabolismo , Dopamina/metabolismo , Ácido Homovanílico/metabolismo , Camundongos , Camundongos Knockout , Microglia/metabolismo , Transtornos Parkinsonianos/genética , Receptor 4 Toll-Like/genética
20.
Vaccine ; 30(33): 4971-6, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22658927

RESUMO

It is estimated that 30-100 million people are infected with the pathogenic nematode Strongyloides stercoralis worldwide but parasite control is still based on anti-helminthic treatment. To develop protective vaccination strategies, we use the murine model of Strongyloides ratti infection. We have shown recently that vaccination with alum-precipitated, but not with native or CFA-emulsified S. ratti heat shock protein 60 (srHSP60) conferred protection to challenge infection. Here we describe the generation of a monoclonal IgM specific for srHSP60. Anti-srHSP60 detected human and srHSP60 and stained S. ratti infective larvae in vitro. Passive immunization of mice with monoclonal anti-srHSP60 IgM led to reduced numbers of migrating larvae in lung and head, reduced numbers of parasitic adults in the small intestine and reduced larval output upon S. ratti challenge infection. Taken together, our findings highlight the relevance of srHSP60 as vaccine candidate for the induction of antibody-mediated protection against Strongyloides infection.


Assuntos
Anticorpos Anti-Helmínticos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Imunização Passiva/métodos , Imunoglobulina M/administração & dosagem , Estrongiloidíase/prevenção & controle , Animais , Anticorpos Anti-Helmínticos/imunologia , Anticorpos Monoclonais/imunologia , Antígenos de Helmintos/imunologia , Chaperonina 60/imunologia , Modelos Animais de Doenças , Fezes/parasitologia , Feminino , Imunoglobulina M/imunologia , Intestino Delgado/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Carga Parasitária , Strongyloides ratti/imunologia , Estrongiloidíase/imunologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA