Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Alzheimers Dement ; 20(1): 253-265, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37578203

RESUMO

INTRODUCTION: Multiple infectious agents, including viruses, bacteria, fungi, and protozoa, have been linked to Alzheimer's disease (AD) risk by independent lines of evidence. We explored this association by comparing the frequencies of viral species identified in a large sample of AD cases and controls. METHODS: DNA sequence reads that did not align to the human genome in sequences were mapped to viral reference sequences, quantified, and then were tested for association with AD in whole exome sequences (WES) and whole genome sequences (WGS) datasets. RESULTS: Several viruses were significant predictors of AD according to the machine learning classifiers. Subsequent regression analyses showed that herpes simplex type 1 (HSV-1) (odds ratio [OR] = 3.71, p = 8.03 × 10-4) and human papillomavirus 71 (HPV-71; OR = 3.56, p = 0.02), were significantly associated with AD after Bonferroni correction. The phylogenetic-related cluster of Herpesviridae was significantly associated with AD in several strata of the data (p < 0.01). DISCUSSION: Our results support the hypothesis that viral infection, especially HSV-1, is associated with AD risk.


Assuntos
Doença de Alzheimer , Herpes Simples , Herpesvirus Humano 1 , Humanos , Doença de Alzheimer/complicações , Filogenia , Herpesvirus Humano 1/genética , DNA
2.
Front Immunol ; 12: 624197, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33815376

RESUMO

Vaccines have played a pivotal role in improving public health, however, many infectious diseases lack an effective vaccine. Controlling the spread of infectious diseases requires continuing studies to develop new and improved vaccines. Our laboratory has been investigating the immune enhancing mechanisms of Toll-like receptor (TLR) ligand-based adjuvants, including the TLR2 ligand Neisseria meningitidis outer membrane protein, PorB. Adjuvant use of PorB increases costimulatory factors on antigen presenting cells (APC), increases antigen specific antibody production, and cytokine producing T cells. We have demonstrated that macrophage expression of MyD88 (required for TLR2 signaling) is an absolute requirement for the improved antibody response induced by PorB. Here-in, we specifically investigated the role of subcapsular CD169+ marginal zone macrophages in antibody production induced by the use of TLR-ligand based adjuvants (PorB and CpG) and non-TLR-ligand adjuvants (aluminum salts). CD169 knockout mice and mice treated with low dose clodronate treated animals (which only remove marginal zone macrophages), were used to investigate the role of these macrophages in adjuvant-dependent antibody production. In both sets of mice, total antigen specific immunoglobulins (IgGs) were diminished regardless of adjuvant used. However, the greatest reduction was seen with the use of TLR ligands as adjuvants. In addition, the effect of the absence of CD169+ macrophages on adjuvant induced antigen and antigen presenting cell trafficking to the lymph nodes was examined using immunofluorescence by determining the relative extent of antigen loading on dendritic cells (DCs) and antigen deposition on follicular dendritic cells (FDC). Interestingly, only vaccine preparations containing PorB had significant decreases in antigen deposition in lymphoid follicles and germinal centers in CD169 knockout mice or mice treated with low dose clodronate as compared to wildtype controls. Mice immunized with CpG containing preparations demonstrated decreased FDC networks in the mice treated with low dose clodronate. Conversely, alum containing preparations only demonstrated significant decreases in IgG in CD169 knockout mice. These studies stress that importance of subcapsular macrophages and their unique role in adjuvant-mediated antibody production, potentially due to an effect of these adjuvants on antigen trafficking to the lymph node and deposition on follicular dendritic cells.


Assuntos
Adjuvantes Imunológicos/farmacologia , Compostos de Alúmen/farmacologia , Imunogenicidade da Vacina , Macrófagos/efeitos dos fármacos , Oligodesoxirribonucleotídeos/farmacologia , Ovalbumina/farmacologia , Porinas/farmacologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Receptores Toll-Like/agonistas , Animais , Ácido Clodrônico/farmacologia , Células Dendríticas Foliculares/efeitos dos fármacos , Células Dendríticas Foliculares/imunologia , Células Dendríticas Foliculares/metabolismo , Imunoglobulina G/sangue , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodesoxirribonucleotídeos/imunologia , Ovalbumina/imunologia , Porinas/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Transdução de Sinais , Receptores Toll-Like/metabolismo , Vacinação
3.
Int J Infect Dis ; 99: 28-33, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32721528

RESUMO

OBJECTIVE: The aim of this observational study was to determine the optimal timing of interleukin-6 receptor inhibitor (IL6ri) administration for coronavirus disease 2019 (COVID-19). METHODS: Patients with COVID-19 were given an IL6ri (sarilumab or tocilizumab) based on iteratively reviewed guidelines. IL6ri were initially reserved for critically ill patients, but after review, treatment was liberalized to patients with lower oxygen requirements. Patients were divided into two groups: those requiring ≤45% fraction of inspired oxygen (FiO2) (termed stage IIB) and those requiring >45% FiO2 (termed stage III) at the time of IL6ri administration. The main outcomes were all-cause mortality, discharge alive from hospital, and extubation. RESULTS: A total of 255 COVID-19 patients were treated with IL6ri (149 stage IIB and 106 stage III). Patients treated in stage IIB had lower mortality than those treated in stage III (adjusted hazard ratio (aHR) 0.24, 95% confidence interval (CI) 0.08-0.74). Overall, 218 (85.5%) patients were discharged alive. Patients treated in stage IIB were more likely to be discharged (aHR 1.43, 95% CI 1.06-1.93) and were less likely to be intubated (aHR 0.43, 95% CI 0.24-0.79). CONCLUSIONS: IL6ri administration prior to >45% FiO2 requirement was associated with improved COVID-19 outcomes. This can guide clinical management pending results from randomized controlled trials.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Betacoronavirus , Infecções por Coronavirus/tratamento farmacológico , Interleucina-6/antagonistas & inibidores , Pneumonia Viral/tratamento farmacológico , COVID-19 , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/patologia , Feminino , Humanos , Intubação Intratraqueal , Masculino , Pessoa de Meia-Idade , Pandemias , Alta do Paciente , Pneumonia Viral/mortalidade , Pneumonia Viral/patologia , SARS-CoV-2 , Resultado do Tratamento
4.
Vaccine ; 36(50): 7689-7699, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30381152

RESUMO

Vaccines formulated with adjuvant have been effective against numerous infectious diseases, almost always due to induction of functional antibodies that recognizes the pathogen of interest. There is an unmet clinical need for vaccine adjuvants that induce T cells responses to potentially enhance protection against malignancies and intracellular pathogens, where a humoral response, alone, may not be adequate for protection. In this study, we demonstrate that a TLR2 ligand-based adjuvant, meningococcal PorB, has broad immunostimulatory activity with the ability to induce a robust and diverse vaccine antigen specific T cell response. We demonstrate that a vaccine formulated with PorB admixed with ovalbumin induces a wide variety of antigen specific antibody subclasses and effector molecules (MIG, MCP-1, IP-10, MIP-1α, KC & IL-2) with known roles for inducing T cell responses, along with elevated levels of Th1 and Th2 type cytokines upon antigen stimulation. We confirmed production of these cytokines by examining the antigen-specific T cells induced by PorB in vivo. After two immunizations with vaccine formulated with PorB/OVA, antigen-specific CD4 and CD8 T cells were significantly increased in numbers and produced IL-4 or IFN-γ upon ex vivo antigen re-stimulation. Finally, in a Listeria mouse infection model, vaccine formulated with PorB significantly reduced the bacterial burden upon a low dose infection and increased survival upon a high dose infection with recombinant Listeria monocytogenes engineered to express OVA (rLmOVA), a pathogen that requires OVA-antigen specific cytotoxic CD8 T cells for clearance. In summary, PorB is able to induce antigen specific broad B and T cell responses, illustrating its potential as a potent and new vaccine adjuvant.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Imunidade Celular , Ovalbumina/imunologia , Porinas/administração & dosagem , Linfócitos T/imunologia , Animais , Carga Bacteriana , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Citocinas/análise , Modelos Animais de Doenças , Listeria monocytogenes/isolamento & purificação , Listeriose/patologia , Listeriose/prevenção & controle , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia
5.
Infect Immun ; 86(8)2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29760215

RESUMO

The gonococcal Opa proteins are an antigenically variable family of surface adhesins that bind human carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), CEACAM3, CEACAM5, and/or CEACAM6, cell surface glycoproteins that are differentially expressed on a broad spectrum of human cells and tissues. While they are presumed to be important for infection, the significance of various Opa-CEACAM-mediated cellular interactions in the context of the genital tract has remained unclear. Here, we observed that CEACAM1 and CEACAM5 are differentially expressed on epithelia lining the upper and lower portions of the human female genital tract, respectively. Using transgenic mouse lines expressing human CEACAMs in a manner that reflects this differential pattern, we considered the impact of Opa-CEACAM interactions during uncomplicated lower genital tract infections versus during pelvic inflammatory disease. Our results demonstrate that Opa-CEACAM5 binding on vaginal epithelia facilitates the long-term colonization of the lower genital tract, while Opa protein binding to CEACAM1 on uterine epithelia enhances gonococcal association and penetration into these tissues. While these Opa-dependent interactions with CEACAM-expressing epithelial surfaces promote infection, Opa binding by neutrophil-expressed CEACAMs counterbalances this by facilitating more effective gonococcal clearance. Furthermore, during uterine infections, CEACAM-dependent tissue invasion aggravates disease pathology by increasing the acute inflammatory response. Together, these findings demonstrate that the outcome of infection is determined by both the cell type-specific expression of human CEACAMs and the CEACAM specificity of the Opa variants expressed, which combine to determine the level of gonococcal association with the genital mucosa versus the extent of CEACAM-dependent inflammation and gonococcal clearance by neutrophils.


Assuntos
Antígenos CD/metabolismo , Aderência Bacteriana , Proteínas da Membrana Bacteriana Externa/metabolismo , Antígeno Carcinoembrionário/metabolismo , Moléculas de Adesão Celular/metabolismo , Genitália Feminina/patologia , Gonorreia/fisiopatologia , Infecções do Sistema Genital/fisiopatologia , Animais , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Proteínas Ligadas por GPI/metabolismo , Perfilação da Expressão Gênica , Genitália Feminina/microbiologia , Gonorreia/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Imuno-Histoquímica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neisseria gonorrhoeae/fisiologia , Infecções do Sistema Genital/microbiologia , Resultado do Tratamento , Útero/microbiologia , Útero/patologia , Vagina/microbiologia , Vagina/patologia
6.
Microbes Infect ; 18(10): 627-638, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27373686

RESUMO

Neisseria meningitidis (NM) is an opportunistic gram-negative human pathogen that colonizes the human nasopharyngeal epithelium. Asymptomatic carriage is common, but some meningococcal strains can invade nasopharyngeal epithelial cells and proceed to cause severe and often fatal infections. Invasion is predominantly driven by expression of bacterial virulence factors and host cell cognate receptors for bacterial recognition. Porins are among the Neisserial components involved in host cell activation and bacterial internalization processes. Similar to other virulence factors, porins present antigenic and structure variability among strains. Such sequence variability in the surface-exposed loop regions has been correlated to bacterial invasiveness and to variability in host cell responses via Toll-like receptor 2 (TLR2). Here, we examined whether TLR2 signaling by porins influences recovery of intracellular Neisseriae from epithelial cells in vitro. Our results show that TLR2 stimulation, either by the organism or exogenously, generally enhances Neisseriae internalization by epithelial cells. TLR2-driven intracellular signaling via ERK1/2, JNK and particularly NF-κB plays a role in this process. Based on these results, it is possible that expression of porin sequence variants that strongly induce TLR2 activation may be a mechanism to enhance the invasive features of pathogenic Neisseriae strains.


Assuntos
Endocitose , Células Epiteliais/microbiologia , Interações Hospedeiro-Patógeno , Neisseria meningitidis/fisiologia , Porinas/metabolismo , Receptor 2 Toll-Like/metabolismo , Fatores de Virulência/metabolismo , Células Cultivadas , Humanos , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo
7.
ACS Nano ; 10(1): 1189-200, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26720275

RESUMO

Membrane-wrapped nanoparticles represent a versatile platform for utilizing specific lipid-receptor interactions, such as siallyllactose-mediated binding of the ganglioside GM3 to Siglec1 (CD169), for targeting purposes. The membrane wrap around the nanoparticles not only serves as a matrix to incorporate GM3 as targeting moiety for antigen-presenting cells but also offers unique opportunities for constructing a biomimetic surface from lipids with potentially protein-repellent properties. We characterize nonspecific protein adsorption (corona formation) to membrane-wrapped nanoparticles with core diameters of approximately 35 and 80 nm and its effect on the GM3-mediated targeting efficacy as a function of surface charge through combined in vitro and in vivo studies. The stability and fate of the membrane wrap around the nanoparticles in a simulated biological fluid and after uptake in CD169-expressing antigen-presenting cells is experimentally tested. Finally, we demonstrate in hock immunization studies in mice that GM3-decorated membrane-wrapped nanoparticles achieve a selective enrichment in the peripheral regions of popliteal lymph nodes that contain high concentrations of CD169-expressing antigen-presenting cells.


Assuntos
Células Dendríticas/efeitos dos fármacos , Gangliosídeo G(M3)/administração & dosagem , Lipossomos/administração & dosagem , Linfonodos/imunologia , Nanopartículas/administração & dosagem , 1,2-Dipalmitoilfosfatidilcolina/análogos & derivados , 1,2-Dipalmitoilfosfatidilcolina/química , Animais , Materiais Biomiméticos/química , Linhagem Celular , Células Dendríticas/citologia , Células Dendríticas/imunologia , Gangliosídeo G(M3)/química , Gangliosídeo G(M3)/imunologia , Expressão Gênica , Células HeLa , Humanos , Imunização , Lipossomos/química , Lipossomos/imunologia , Linfonodos/citologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Nanopartículas/química , Fosfatidilserinas/química , Cultura Primária de Células , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Propriedades de Superfície , Vírion/química , Vírion/imunologia
8.
PLoS One ; 8(12): e82171, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349212

RESUMO

Vaccines play a vital role in modern medicine. The development of novel vaccines for emerging and resistant pathogens has been aided in recent years by the use of novel adjuvants in subunit vaccines. A deeper understanding of the molecular pathways behind adjuvanticity is required to better select immunostimulatory molecules for use in individual vaccines. To this end, we have undertaken a study of the essential signaling pathways involved in the innate and adaptive immune responses to the Neisseria meningitidis outer membrane protein Porin B (PorB). We have previously demonstrated that PorB is an agonist of Toll-Like Receptor 2 (TLR2) and acts as an adjuvant in vaccines for protein, carbohydrate and lipopolysaccharide antigens using murine models. Here we demonstrate NFκB translocation following stimulation with PorB only occurs in the presence of TLR2. IL-6 and TNF-α secretion was shown to be MAPK dependent. Surface expression of activation markers on macrophages, including CD40, CD69, and CD86, was increased following PorB stimulation in vitro. Interestingly, some upregulation of CD54 and CD69 was still observed in macrophages obtained from TLR2 KO mice, indicating a possible non-TLR2 mediated activation pathway induced by PorB. In a murine vaccination model, using ovalbumin as the antigen and PorB as the adjuvant, a decreased antigen-specific IgG production was observed in TLR2 KO mice; adjuvant-dependent increased IgG production was entirely ablated in MyD88 KO mice. These observations demonstrate the importance of the above pathways to the adjuvant activity of PorB. The potential TLR2 independent effect is currently being explored.


Assuntos
Imunidade/efeitos dos fármacos , Porinas/farmacologia , Imunidade Adaptativa/efeitos dos fármacos , Trifosfato de Adenosina/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citocinas/metabolismo , Feminino , Imunoglobulina G/metabolismo , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-1beta/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 2 Toll-Like/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
J Immunol ; 190(3): 1148-57, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23264656

RESUMO

Porphyromonas gingivalis is a primary etiological agent of chronic periodontal disease, an infection-driven chronic inflammatory disease that leads to the resorption of tooth-supporting alveolar bone. We previously reported that TLR2 is required for P. gingivalis-induced alveolar bone loss in vivo, and our in vitro work implicated TNF as a key downstream mediator. In this study, we show that TNF-deficient (Tnf(-/-)) mice are resistant to alveolar bone loss following oral infection with P. gingivalis, and thus establish a central role for TNF in experimental periodontal disease. Using bone marrow-derived macrophages (BMDM) from wild-type and gene-specific knockout mice, we demonstrate that the initial inflammatory response to P. gingivalis in naive macrophages is MyD88 dependent and requires cooperative signaling of TLR2 and TLR4. The ability of P. gingivalis to activate cells via TLR2 or TLR4 was confirmed in TLR2- or TLR4-transformed human embryonic kidney cells. Additional studies using bacterial mutants demonstrated a role for fimbriae in the modulation of TLR-mediated activation of NF-κB. Whereas both TLR2 and TLR4 contributed to TNF production in naive macrophages, P. gingivalis preferentially exploited TLR2 in endotoxin-tolerant BMDM to trigger excessive TNF production. We found that TNF induced surface TLR2 expression and augmented TLR-induced cytokine production in P. gingivalis-stimulated BMDM, establishing a previously unidentified TNF-dependent feedback loop. Adoptive transfer of TLR2-expressing macrophages to TLR2-deficient mice restored the ability of P. gingivalis to induce alveolar bone loss in vivo. Collectively, our results identify a TLR2- and TNF-dependent macrophage-specific mechanism underlying pathogen-induced inflammatory bone loss in vivo.


Assuntos
Perda do Osso Alveolar/etiologia , Infecções por Bacteroidaceae/imunologia , Gengivite/fisiopatologia , Macrófagos/fisiologia , Porphyromonas gingivalis/patogenicidade , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Transferência Adotiva , Perda do Osso Alveolar/imunologia , Perda do Osso Alveolar/fisiopatologia , Animais , Antibioticoprofilaxia , Infecções por Bacteroidaceae/microbiologia , Fímbrias Bacterianas/fisiologia , Regulação da Expressão Gênica/imunologia , Gengivite/complicações , Gengivite/imunologia , Células HEK293 , Humanos , Teste de Cultura Mista de Linfócitos , Ativação de Macrófagos , Macrófagos/transplante , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , NF-kappa B/metabolismo , Porphyromonas gingivalis/genética , Porphyromonas gingivalis/imunologia , Porphyromonas gingivalis/ultraestrutura , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Fator de Necrose Tumoral alfa/deficiência
10.
Clin Immunol ; 138(1): 33-40, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20947433

RESUMO

There is a need for developing vaccines that elicit mucosal immunity. Although oral or nasal vaccination methods would be ideal, current strategies have yielded mixed success. Toll-like receptor 2 (TLR2) ligands are effective adjuvants and are currently used in the Haemophilus influenzae type B vaccine. Induction of humoral immunity in the mucosa is critical for effective vaccination; thus, we sought to determine the effects of TLR2 ligands on human mucosal B cell differentiation. We demonstrate that TLR2 ligands induce CCR9 and CCR10 expression by circulating B cells and increased chemotaxis to cognate chemokines CCL25 and CCL28 suggesting that TLR2 induces B cell homing to the gastrointestinal tract. TLR2 stimulation of B cells also induced J chain and IgA production demonstrating the induction of mucosal-like antibody secreting cells. These observations suggest that vaccines containing TLR2-ligands as adjuvants could induce mucosal B cell immunity even when delivered in a non-mucosal manner.


Assuntos
Linfócitos B/metabolismo , Imunidade nas Mucosas/imunologia , Imunoglobulina A/biossíntese , Receptores de Retorno de Linfócitos/metabolismo , Receptor 2 Toll-Like/metabolismo , Adolescente , Adulto , Idoso , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Quimiocinas CC/farmacologia , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/imunologia , Doença de Crohn/imunologia , Doença de Crohn/patologia , Profilaxia Dentária , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Íleo/imunologia , Íleo/metabolismo , Íleo/patologia , Cadeias J de Imunoglobulina/metabolismo , Imunoglobulina M/metabolismo , Doenças Inflamatórias Intestinais/imunologia , Interleucina-10/farmacologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Lipopolissacarídeos/sangue , Lipopolissacarídeos/farmacologia , Masculino , Pessoa de Meia-Idade , Tonsila Palatina/citologia , Tonsila Palatina/imunologia , Porinas/imunologia , Porinas/farmacologia , Receptores CCR/genética , Receptores CCR/metabolismo , Receptores CCR10/genética , Receptores CCR10/metabolismo , Adulto Jovem
11.
Infect Immun ; 78(3): 994-1003, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20028813

RESUMO

Meningococcal porin PorB is an inhibitor of apoptosis induced via the intrinsic pathway in various cell types. This effect is attributed to prevention of mitochondrial depolarization and of subsequent release of proapoptotic mitochondrial factors. To determine whether apoptosis is globally inhibited by PorB, we compared the intrinsic and extrinsic pathways in HeLa cells. Interestingly, PorB does not prevent extrinsic apoptosis induced by tumor necrosis factor alpha plus cycloheximide, suggesting a unique mitochondrial pathway specificity. Several intracellular factors regulated by NF-kappaB, including members of the Bcl-2 family and of the inhibitor of apoptosis (IAP) family, play major roles in controlling apoptosis, and some of them are thought to contribute to the antiapoptotic effect of the gonococcal porin, PIB. However, most of the members of the Bcl-2 family and the IAP family are not induced by meningococcal PorB in HeLa cells, with the exception of Bfl-1/A1. Interestingly, PorB does not induce NF-kappaB activation in HeLa cells, likely due to a lack of Toll-like receptor 2 (TLR2) expression in these cells. Bfl-1/A1 expression is also regulated by CBF1, a nuclear component of the Notch signaling pathway, independent of NF-kappaB activation. Since HeLa cells are protected by PorB from intrinsic apoptosis events, regardless of TLR2 and NF-kappaB expression, the possibility of a contribution of alternative signaling pathways to this effect cannot be excluded. In this paper, we describe an initial dissection of the cascade of cellular events involved in the antiapoptotic effect of PorB in the absence of TLR2.


Assuntos
Apoptose , NF-kappa B/imunologia , Neisseria meningitidis/imunologia , Neisseria meningitidis/patogenicidade , Porinas/fisiologia , Receptor 2 Toll-Like/imunologia , Fatores de Virulência/fisiologia , Células Epiteliais/microbiologia , Células HeLa , Humanos
12.
Infect Immun ; 76(2): 486-96, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18025095

RESUMO

Francisella tularensis can cause severe disseminated disease after respiratory infection. The identification of factors involved in mortality or recovery following induction of tularemia in the mouse will improve our understanding of the natural history of this disease and facilitate future evaluation of vaccine candidate preparations. BALB/c mice were infected intranasally with the live vaccine strain (LVS) of F. tularensis subsp. holarctica and euthanized at different stages of disease to analyze the induction of immune molecules, gross anatomical features of organs, bacterial burdens, and progression of the histopathological changes in lung and spleen. Tissue-specific interleukin-6 (IL-6), macrophage inflammatory protein 2, and monocyte chemotactic protein 1 were immune markers of mortality, while anti-LVS immunoglobulin M and IL-1beta were associated with survival. Moribund mice had enlarged spleens and lungs, while surviving mice had even more prominent splenomegaly and normal-appearing lungs. Histopathology of the spleens of severely ill mice was characterized by disrupted lymphoid follicles and fragmented nuclei, while the spleens of survivors appeared healthy but with increased numbers of megakaryocytes and erythrocytes. Histopathology of the lungs of severely ill mice indicated severe pneumonia. Lungs of survivors at early time points showed increased inflammation, while at late times they appeared healthy with peribronchial lymphoid aggregates. Our results suggest that host immune factors are able to affect bacterial dissemination after respiratory tularemia, provide new insights regarding the pathological characteristics of pulmonary tularemia leading to systemic disease, and potentially identify immune markers associated with recovery from the disease.


Assuntos
Francisella tularensis/imunologia , Pneumonia/imunologia , Pneumonia/patologia , Tularemia/imunologia , Tularemia/patologia , Animais , Anticorpos Antibacterianos/análise , Peso Corporal , Quimiocina CCL2/análise , Quimiocina CXCL2/análise , Contagem de Colônia Microbiana , Feminino , Imunoglobulina M/análise , Interleucina-1beta/análise , Interleucina-6/análise , Pulmão/química , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Pneumonia/microbiologia , Baço/química , Baço/microbiologia , Baço/patologia
13.
Cell Microbiol ; 7(12): 1736-48, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16309460

RESUMO

Purified pili and porin from Neisseria quickly mobilize calcium (Ca(2+)) stores in monocytes and epithelial cells, ultimately influencing host cell viability as well as bacterial intracellular survival. Here, we examined the Ca(2+) transients induced in human epithelial cells during infection by live, piliated N. gonorrhoeae. Porin induced an influx of Ca(2+) from the extracellular medium less than 60 s post infection. The porin-induced transient is followed by a pilus-induced release of Ca(2+) from intracellular stores. The timing of these events is similar to that observed using purified proteins. Interestingly, the porin-induced Ca(2+) flux is required for the pilus-induced transient, indicating that the pilus-induced Ca(2+) release is, itself, Ca(2+) dependent. Several lines of evidence indicate that porin is present on pili. Moreover, pilus retraction strongly influences the porin- and pilus-induced Ca(2+) fluxes. These and other results strongly suggest that the pilus and porin cooperate to modulate calcium signalling in epithelial cells, and propose a model to explain how N. gonorrhoeae triggers Ca(2+) transients in the initial stages of pilus-mediated attachment.


Assuntos
Cálcio/metabolismo , Fímbrias Bacterianas/metabolismo , Neisseria gonorrhoeae/metabolismo , Porinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Humanos , Transdução de Sinais
14.
J Immunol ; 175(9): 5596-600, 2005 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16237045

RESUMO

MyD88 is an adaptor protein for the TLR family of proteins that has been implicated as a critical mediator of innate immune responses to pathogen detection. In this study, we report that MyD88 plays a crucial role in killing Gram-negative bacteria by primary macrophages via influencing NADPH oxidase function. Peritoneal macrophages from MyD88-/- mice exhibited a marked inability to kill Escherichia coli (F18) or an attenuated strain of Salmonella typhimurium (sseB) in vitro. This defect in killing was due to diminished NADPH oxidase-mediated production of superoxide anion in response to bacteria by MyD88-/- phagocytes as a consequence of defective NADPH oxidase assembly. Defective oxidase assembly in MyD88-deficient macrophages resulted from impaired p38 MAPK activation and subsequent phosphorylation of p47phox. Together these data demonstrate a pivotal role for MyD88 in killing Gram-negative bacteria via modulation of NADPH oxidase activity in phagocytic cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Antígenos de Diferenciação/fisiologia , Bactérias Gram-Negativas/imunologia , Macrófagos/enzimologia , Macrófagos/imunologia , NADPH Oxidases/fisiologia , Fagocitose , Receptores Imunológicos/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
15.
J Immunol ; 174(6): 3545-50, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15749891

RESUMO

Neisserial porins have been shown to act as B cell mitogens and immune adjuvants. PorA and PorB are the major outer membrane porin proteins of the human pathogen Neisseria meningitidis. We have shown that the mechanism of the immunopotentiating capability of porin involves up-regulation of the T cell costimulatory ligand, CD86. Due to neisserial porin's ability to activate B cells and potentiate immune responses, we hypothesized that porin also employs the potent immune stimulatory function of dendritic cells (DC). We examined the ability of purified N. meningitidis PorB to induce maturation of murine splenic and bone marrow-derived DC. PorB treatment induced DC maturation, as demonstrated by increased expression of CD86 and class I and II MHC molecules. In addition, PorB not only enhanced the allostimulatory activity of DC, but also augmented the ability of DC to stimulate T cells in an Ag-specific manner. PorB-matured DC secreted the inflammatory cytokine IL-6, which may have implications for the adjuvant property of porin. Induction of IL-6 by PorB is also significant because IL-6 is one of a number of cytokines produced during infection with N. meningitidis and may be involved in the inflammatory process observed during infection and disease. We previously demonstrated the requirement of MyD88 and TLR2 for PorB-induced B cell activation. In the present study, MyD88 and TLR2 were also essential for PorB-induced DC activation. This work is significant for elucidating the mechanism(s) of neisserial porin's immune stimulatory activity.


Assuntos
Antígenos de Diferenciação/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Neisseria meningitidis/imunologia , Porinas/farmacologia , Receptores de Superfície Celular/metabolismo , Receptores Imunológicos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação/genética , Antígeno B7-2 , Feminino , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Fatores Imunológicos/farmacologia , Interleucina-6/biossíntese , Ativação Linfocitária , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide , Neisseria meningitidis/patogenicidade , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Linfócitos T/imunologia , Receptor 2 Toll-Like
16.
Infect Immun ; 70(11): 5965-71, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12379671

RESUMO

The immunoglobulin A (IgA) protease secreted by pathogenic Neisseria spp. cleaves Lamp1, thereby altering lysosomes in a cell and promoting bacterial intracellular survival. We sought to determine how the IgA protease gains access to cellular Lamp1 in order to better understand the role of this cleavage event in bacterial infection. In a previous report, we demonstrated that the pilus-induced Ca(2+) transient triggers lysosome exocytosis in human epithelial cells. This, in turn, increases the level of Lamp1 at the plasma membrane, where it can be cleaved by IgA protease. Here, we show that porin also induces a Ca(2+) flux in epithelial cells. This transient is similar in nature to that observed in phagocytes exposed to porin. In contrast to the pilus-induced Ca(2+) transient, the porin-induced event does not trigger lysosome exocytosis. Instead, it stimulates exocytosis of early and late endosomes and increases Lamp1 on the cell surface. These results indicate that Neisseria pili and porin perturb Lamp1 trafficking in epithelial cells by triggering separate and distinct Ca(2+)-dependent exocytic events, bringing Lamp1 to the cell surface, where it can be cleaved by IgA protease.


Assuntos
Antígenos CD/metabolismo , Membrana Celular/metabolismo , Endossomos/metabolismo , Exocitose/efeitos dos fármacos , Neisseria gonorrhoeae/fisiologia , Porinas/farmacologia , Cálcio/metabolismo , Humanos , Proteína 1 de Membrana Associada ao Lisossomo , Proteínas de Membrana Lisossomal , Serina Endopeptidases/fisiologia , Células Tumorais Cultivadas
17.
J Immunol ; 168(4): 1533-7, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11823477

RESUMO

The immunopotentiating activity of neisserial porins, the major outer membrane protein of the pathogenic Neisseria, is mediated by its ability to stimulate B cells and up-regulate the surface expression of B7-2. This ability is dependent on MyD88 and Toll-like receptor (TLR)2 expression, as demonstrated by a lack of a response by B cells from MyD88 or TLR2 knockout mice to the porins. Using previously described TLR2-dependent reporter constructs, these results were confirmed and were shown to be due to induction of NF-kappaB nuclear translocation. This is the first demonstration of known vaccine adjuvant to stimulate immune cells via TLR2.


Assuntos
Antígenos de Diferenciação/fisiologia , Linfócitos B/imunologia , Proteínas de Drosophila , Glicoproteínas de Membrana/fisiologia , Neisseria meningitidis/imunologia , Porinas/imunologia , Receptores de Superfície Celular/fisiologia , Receptores Imunológicos/fisiologia , Transporte Ativo do Núcleo Celular , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Bactérias/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação/genética , Antígeno B7-2 , Células CHO , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Cricetinae , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , NF-kappa B/metabolismo , Receptores de Superfície Celular/genética , Receptores Imunológicos/genética , Receptor 2 Toll-Like , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA