Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Infect Dis Obstet Gynecol ; 2020: 8890619, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33082702

RESUMO

Preterm birth is a major public health problem, occurring in more than half a million births per year in the United States. A number of maternal conditions have been recognized as risk factors for preterm birth, but for the majority of cases, the etiology is not completely understood. Chlamydia trachomatis is one of the most prevalent sexually transmitted infections in the world. However, its role in adverse pregnancy outcome in women is still debated. In order to determine if genitourinary tract infection with C. trachomatis during pregnancy was associated with preterm birth, we conducted a case-control study on women who delivered at Boston Medical Center, an urban "safety-net" hospital that serves a socioeconomically disadvantaged and racially diverse population. Women with known risk factors for preterm birth or immune suppression were excluded. Variables collected on enrolled subjects included demographics; diagnosis of C. trachomatis during or prior to pregnancy; tobacco, alcohol, and illicit substance use; gestational age; and birthweight and gender of the newborn. We also collected urine for chlamydia testing at the time of delivery and placental biopsies for nucleic acid amplification and histological studies. A total of 305 subjects were enrolled: 100 who delivered preterm and 205 who delivered full term. Among those subjects, we identified 19 cases of pregnancy-associated C. trachomatis infection: 6/100 preterm and 13/205 full term, a difference which was not statistically significant. Only two cases of untreated chlamydia infection were identified postpartum, and both occurred in women who delivered at term. We conclude that genitourinary tract infection with C. trachomatis during pregnancy, when appropriately treated, is not associated with preterm birth.


Assuntos
Infecções por Chlamydia/tratamento farmacológico , Chlamydia trachomatis/isolamento & purificação , Complicações Infecciosas na Gravidez/tratamento farmacológico , Nascimento Prematuro/epidemiologia , Adolescente , Adulto , Estudos de Casos e Controles , Infecções por Chlamydia/diagnóstico , Infecções por Chlamydia/epidemiologia , Chlamydia trachomatis/genética , DNA Bacteriano/genética , Feminino , Hospitais Urbanos , Humanos , Idade Materna , Placenta/microbiologia , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/epidemiologia , Fatores de Risco , Provedores de Redes de Segurança , Urina/microbiologia , Adulto Jovem
2.
PLoS Pathog ; 9(8): e1003569, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24009502

RESUMO

The sst1, "supersusceptibility to tuberculosis," locus has previously been shown to be a genetic determinant of host resistance to infection with the intracellular pathogen, Mycobacterium tuberculosis. Chlamydia pneumoniae is an obligate intracellular bacterium associated with community acquired pneumonia, and chronic infection with C. pneumoniae has been linked to asthma and atherosclerosis. C. pneumoniae is a highly adapted pathogen that can productively infect macrophages and inhibit host cell apoptosis. Here we examined the role of sst1 in regulating the host response to infection with C. pneumoniae. Although mice carrying the sst1 susceptible (sst1(S) ) locus were not impaired in their ability to clear the acute infection, they were dramatically less tolerant of the induced immune response, displaying higher clinical scores, more severe lung inflammation, exaggerated macrophage and neutrophil influx, and the development of fibrosis compared to wild type mice. This correlated with increased activated caspase-3 in the lungs of infected sst1(S) mice. Infection of sst1(S) macrophages with C. pneumoniae resulted in a shift in the secreted cytokine profile towards enhanced production of interferon-ß and interleukin-10, and induced apoptotic cell death, which was dependent on secretion of interferon-ß. Intriguingly macrophages from the sst1(S) mice failed to support normal chlamydial growth, resulting in arrested development and failure of the organism to complete its infectious cycle. We conclude that the sst1 locus regulates a shared macrophage-mediated innate defense mechanism against diverse intracellular bacterial pathogens. Its susceptibility allele leads to upregulation of type I interferon pathway, which, in the context of C. pneumoniae, results in decreased tolerance, but not resistance, to the infection. Further dissection of the relationship between type I interferons and host tolerance during infection with intracellular pathogens may provide identification of biomarkers and novel therapeutic targets.


Assuntos
Infecções por Chlamydophila/imunologia , Chlamydophila pneumoniae/imunologia , Loci Gênicos/imunologia , Imunidade Inata/fisiologia , Macrófagos Alveolares/imunologia , Pneumonia Bacteriana/imunologia , Animais , Caspase 3/genética , Caspase 3/imunologia , Infecções por Chlamydophila/genética , Infecções por Chlamydophila/patologia , Chlamydophila pneumoniae/genética , Chlamydophila pneumoniae/ultraestrutura , Evasão da Resposta Imune/genética , Interferon beta/genética , Interferon beta/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Macrófagos Alveolares/ultraestrutura , Camundongos , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/patologia
3.
BMC Genomics ; 15: 1176, 2014 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-25540039

RESUMO

BACKGROUND: Atherosclerosis is a progressive disease characterized by inflammation and accumulation of lipids in vascular tissue. Porphyromonas gingivalis (Pg) and Chlamydia pneumoniae (Cp) are associated with inflammatory atherosclerosis in humans. Similar to endogenous mediators arising from excessive dietary lipids, these Gram-negative pathogens are pro-atherogenic in animal models, although the specific inflammatory/atherogenic pathways induced by these stimuli are not well defined. In this study, we identified gene expression profiles that characterize P. gingivalis, C. pneumoniae, and Western diet (WD) at acute and chronic time points in aortas of Apolipoprotein E (ApoE-/-) mice. RESULTS: At the chronic time point, we observed that P. gingivalis was associated with a high number of unique differentially expressed genes compared to C. pneumoniae or WD. For the top 500 differentially expressed genes unique to each group, we observed a high percentage (76%) that exhibited decreased expression in P. gingivalis-treated mice in contrast to a high percentage (96%) that exhibited increased expression in WD mice. C. pneumoniae treatment resulted in approximately equal numbers of genes that exhibited increased and decreased expression. Gene Set Enrichment Analysis (GSEA) revealed distinct stimuli-associated phenotypes, including decreased expression of mitochondrion, glucose metabolism, and PPAR pathways in response to P. gingivalis but increased expression of mitochondrion, lipid metabolism, carbohydrate and amino acid metabolism, and PPAR pathways in response to C. pneumoniae; WD was associated with increased expression of immune and inflammatory pathways. DAVID analysis of gene clusters identified by two-way ANOVA at acute and chronic time points revealed a set of core genes that exhibited altered expression during the natural progression of atherosclerosis in ApoE-/- mice; these changes were enhanced in P. gingivalis-treated mice but attenuated in C. pneumoniae-treated mice. Notable differences in the expression of genes associated with unstable plaques were also observed among the three pro-atherogenic stimuli. CONCLUSIONS: Despite the common outcome of P. gingivalis, C. pneumoniae, and WD on the induction of vascular inflammation and atherosclerosis, distinct gene signatures and pathways unique to each pro-atherogenic stimulus were identified. Our results suggest that pathogen exposure results in dysregulated cellular responses that may impact plaque progression and regression pathways.


Assuntos
Aorta/metabolismo , Apolipoproteínas E/deficiência , Chlamydophila pneumoniae/fisiologia , Dieta Ocidental/efeitos adversos , Perfilação da Expressão Gênica , Porphyromonas gingivalis/fisiologia , Animais , Aorta/patologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Família Multigênica/genética , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/genética , Placa Aterosclerótica/microbiologia , Placa Aterosclerótica/patologia
4.
J Immunol ; 184(10): 5743-54, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20393140

RESUMO

Chlamydia pneumoniae is a common respiratory pathogen associated with atypical pneumonia, and it has been suggested as a trigger or promoter of several chronic inflammatory conditions, such as asthma and atherosclerosis. The beta form of IL-1 (IL-1beta) is a proinflammatory cytokine released by many cell types and is an important mediator of inflammation during infection. IL-1beta production is a tightly controlled process that includes regulation at multiple levels and typically requires two distinct signals for activation and release. In this study, we investigated the ability of C. pneumoniae to induce IL-1beta secretion. We found that C. pneumoniae was unique among the other Chlamydia species tested in its ability to potently induce secretion of mature IL-1beta from unprimed bone marrow-derived macrophages during a productive infection. TLR2 was required for induction of pro-IL-1beta, whereas the NLRP3/ASC was required for caspase-1 activation and pro-IL-1beta cleavage to produce mature IL-1beta. Caspase-1 cleavage was independent of endogenous ATP release, but required potassium flux, lysosomal acidification, and cathepsin B release. We further investigated the role of IL-1 in host defense against C. pneumoniae-induced pneumonia using mice deficient in the type I IL-1R. Although the IL-1R(-/-) mice developed an inflammatory infiltrate, the number of infiltrating neutrophils was lower, whereas there was evidence of increased infiltrating fibroblasts and mesenchymal cells and more lung fibrosis. We conclude that C. pneumoniae directly activates the NLRP3/ASC inflammasome, leading to the release of biologically active IL-1beta, and that concurrent IL-1 signaling is required for optimal host defense against acute bacterial pneumonia.


Assuntos
Proteínas de Transporte/fisiologia , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/patologia , Chlamydophila pneumoniae/imunologia , Proteínas do Citoesqueleto/fisiologia , Mediadores da Inflamação/fisiologia , Interleucina-1beta/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/microbiologia , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Infecções por Chlamydia/metabolismo , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/metabolismo , Fibrose , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia , Transdução de Sinais/imunologia
5.
Innate Immun ; 24(1): 66-78, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29297237

RESUMO

Complement is an important arm of the innate immune system. Recent studies have shown that products of complement pathway activation can interact directly with other innate immune signaling molecules, including TLRs and inflammasome family members, during some infectious and chronic inflammatory disorders. Activation of the complement system generates anaphylatoxins, such as C3a and C5a, which modulate inflammation. However, the biological effects of interactions between the anaphylatoxins with their receptors may vary across species. In this study, we demonstrate that human complement and rat complement differ in the way they modulate the inflammatory response to the human pathogen, Neisseria gonorrhoeae, as well as purified pathogen-associated ligands, such as LPS. While rat serum down-regulates MyD88-dependent pro-inflammatory cytokine responses in macrophages, human serum has no effect, or in some cases an enhancing effect. Further, the inhibitory effect of rat serum on otherwise pro-inflammatory stimuli is mediated by complement, specifically C3a-C3a receptor interactions, via an undefined signaling mechanism that down-regulates the transcription factor, NF-κB and NLRP3 inflammasome-mediated caspase-1 activation. This study highlights important functional differences between rodent and human complement that could explain some of the differences in immune responses between these two species. Understanding the crosstalk between complement and other arms of the innate immune system will facilitate the development of better anti-inflammatory therapeutics.


Assuntos
Complemento C3a/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Animais , Ativação do Complemento , Citocinas/biossíntese , Gonorreia/imunologia , Humanos , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/imunologia , NF-kappa B/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/biossíntese , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Neisseria gonorrhoeae/imunologia , Ratos Sprague-Dawley , Transdução de Sinais/imunologia , Especificidade da Espécie
6.
Pathog Dis ; 74(6)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27481727

RESUMO

Macrophage foam cell formation is a key event in atherosclerosis. Several triggers induce low-density lipoprotein (LDL) uptake by macrophages to create foam cells, including infections with Porphyromonas gingivalis and Chlamydia pneumoniae, two pathogens that have been linked to atherosclerosis. While gene regulation during foam cell formation has been examined, comparative investigations to identify shared and specific pathogen-elicited molecular events relevant to foam cell formation are not well documented. We infected mouse bone marrow-derived macrophages with P. gingivalis or C. pneumoniae in the presence of LDL to induce foam cell formation, and examined gene expression using an atherosclerosis pathway targeted plate array. We found over 30 genes were significantly induced in response to both pathogens, including PPAR family members that are broadly important in atherosclerosis and matrix remodeling genes that may play a role in plaque development and stability. Six genes mainly involved in lipid transport were significantly downregulated. The response overall was remarkably similar and few genes were regulated in a pathogen-specific manner. Despite very divergent lifestyles, P. gingivalis and C. pneumoniae activate similar gene expression profiles during foam cell formation that may ultimately serve as targets for modulating infection-elicited foam cell burden, and progression of atherosclerosis.


Assuntos
Células Espumosas/metabolismo , Células Espumosas/patologia , Interações Hospedeiro-Patógeno , Macrófagos/metabolismo , Macrófagos/patologia , Transdução de Sinais , Animais , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Chlamydophila pneumoniae/imunologia , Análise por Conglomerados , Biologia Computacional/métodos , Citocinas/metabolismo , Células Espumosas/imunologia , Células Espumosas/microbiologia , Perfilação da Expressão Gênica , Ontologia Genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Mediadores da Inflamação/metabolismo , Metabolismo dos Lipídeos , Peroxidação de Lipídeos , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Anotação de Sequência Molecular , Porphyromonas gingivalis/imunologia
7.
PLoS One ; 10(7): e0131688, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26148065

RESUMO

INTRODUCTION: Diverse and multi-factorial processes contribute to the progression of cardiovascular disease. These processes affect cells involved in the development of this disease in varying ways, ultimately leading to atherothrombosis. The goal of our study was to compare the differential effects of specific stimuli--two bacterial infections and a Western diet--on platelet responses in ApoE-/- mice, specifically examining inflammatory function and gene expression. Results from murine studies were verified using platelets from participants of the Framingham Heart Study (FHS; n = 1819 participants). METHODS: Blood and spleen samples were collected at weeks 1 and 9 from ApoE-/- mice infected with Porphyromonas gingivalis or Chlamydia pneumoniae and from mice fed a Western diet for 9 weeks. Transcripts based on data from a Western diet in ApoE-/- mice were measured in platelet samples from FHS using high throughput qRT-PCR. RESULTS: At week 1, both bacterial infections increased circulating platelet-neutrophil aggregates. At week 9, these cells individually localized to the spleen, while Western diet resulted in increased platelet-neutrophil aggregates in the spleen only. Microarray analysis of platelet RNA from infected or Western diet-fed mice at week 1 and 9 showed differential profiles. Genes, such as Serpina1a, Ttr, Fgg, Rpl21, and Alb, were uniquely affected by infection and diet. Results were reinforced in platelets obtained from participants of the FHS. CONCLUSION: Using both human studies and animal models, results demonstrate that variable sources of inflammatory stimuli have the ability to influence the platelet phenotype in distinct ways, indicative of the diverse function of platelets in thrombosis, hemostasis, and immunity.


Assuntos
Plaquetas/patologia , Dieta Ocidental/efeitos adversos , Inflamação/patologia , Agregação Plaquetária/fisiologia , Animais , Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Plaquetas/metabolismo , Plaquetas/microbiologia , Chlamydophila pneumoniae/patogenicidade , Modelos Animais de Doenças , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Masculino , Camundongos , Neutrófilos/microbiologia , Neutrófilos/patologia , Neutrófilos/fisiologia , Porphyromonas gingivalis/patogenicidade , Trombose/metabolismo , Trombose/patologia
8.
Innate Immun ; 20(4): 377-89, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-23884094

RESUMO

NOD1 and NOD2 are members of the NOD-like receptor family of cytosolic pattern recognition receptors that recognize specific fragments of the bacterial cell wall component peptidoglycan. Neisseria species are unique amongst Gram-negative bacteria in that they turn over large amounts of peptidoglycan during growth. We examined the ability of NOD1 and NOD2 to recognize Neisseria gonorrhoeae, and determined the role of NOD-dependent signaling in regulating the immune response to gonococcal infection. Gonococci, as well as conditioned medium from mid-logarithmic phase grown bacteria, were capable of activating both human NOD1 and NOD2, as well as mouse NOD2, leading to the activation of the transcription factor NF-κB and polyubiquitination of the adaptor receptor-interacting serine-threonine kinase 2. We identified a number of cytokines and chemokines that were differentially expressed in wild type versus NOD2-deficient macrophages in response to gonococcal infection. Moreover, NOD2 signaling up-regulated complement pathway components and cytosolic nucleic acid sensors, suggesting a broad impact of NOD activation on innate immunity. Thus, NOD1 and NOD2 are important intracellular regulators of the immune response to infection with N. gonorrhoeae. Given the intracellular lifestyle of this pathogen, we believe these cytosolic receptors may provide a key innate immune defense mechanism for the host during gonococcal infection.


Assuntos
Gonorreia/imunologia , Macrófagos/fisiologia , Neisseria gonorrhoeae/imunologia , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Animais , Ativação do Complemento/genética , Células HEK293 , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/genética , Imunomodulação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , Peptidoglicano/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética
9.
PLoS One ; 6(6): e20846, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21695078

RESUMO

Chlamydia trachomatis is a common sexually transmitted pathogen and is associated with infant pneumonia. Data from the female mouse model of genital tract chlamydia infection suggests a requirement for TLR2-dependent signaling in the induction of inflammation and oviduct pathology. We hypothesized that the role of TLR2 in moderating mucosal inflammation is site specific. In order to investigate this, we infected mice via the intranasal route with C. muridarum and observed that in the absence of TLR2 activation, mice had more severe disease, higher lung cytokine levels, and an exaggerated influx of neutrophils and T-cells into the lungs. This could not be explained by impaired bacterial clearance as TLR2-deficient mice cleared the infection similar to controls. These data suggest that TLR2 has an anti-inflammatory function in the lung during Chlamydia infection, and that the role of TLR2 in mucosal inflammation varies at different mucosal surfaces.


Assuntos
Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/microbiologia , Chlamydia muridarum/patogenicidade , Infecções Respiratórias/metabolismo , Infecções Respiratórias/microbiologia , Receptor 2 Toll-Like/metabolismo , Animais , Chlamydia muridarum/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Inflamação/metabolismo , Pulmão/metabolismo , Pulmão/microbiologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Plasmídeos/genética , Receptor 2 Toll-Like/deficiência
10.
J Reprod Immunol ; 84(1): 16-23, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19963275

RESUMO

TNF-like weak inducer of apoptosis (TWEAK) is a member of the tumor necrosis factor (TNF) cytokine superfamily which regulates a number of cellular responses, including inflammation and proliferation. TWEAK is primarily secreted by phagocytic cells and its receptor, fibroblast growth factor-inducible 14 (Fn14), is expressed on non-lymphoid cells, including epithelial, endothelial and mesenchymal cells. The TWEAK/Fn14 pathway is highly conserved from an evolutionary standpoint, and has been shown to play a role in tissue regeneration and inflammation in the liver, kidney, lung and skeletal muscle. We hypothesized that TWEAK/Fn14 might have a physiological role in regulating infection-induced inflammation in the lower female genital tract. To test this hypothesis, we examined expression of the receptor Fn14 in relevant cells and tissue. Receptor function was tested by treating cells with recombinant TWEAK, with and without other known proinflammatory stimuli. Flow cytometric analysis of vaginal and cervical epithelial cells revealed that Fn14 was highly expressed at the cell surface. We also detected both Fn14 and TWEAK in whole cervical tissue by RT-PCR. Treatment of vaginal and cervical epithelial cells with recombinant TWEAK led to a weak induction of the chemokine IL-8. However, TWEAK potentiated the effects of IL-1ss, the TLR2 ligand Pam(3)CysSK(4), and live Neisseria gonorrhoeae in a synergistic manner. These data reveal a novel pathway for regulation of microbial-induced inflammation in the female reproductive tract and suggest that interference with the TWEAK/Fn14 pathway might be an approach to abrogate excessive infection-induced inflammation caused by sexually transmitted pathogens.


Assuntos
Colo do Útero/imunologia , Doenças dos Genitais Femininos/microbiologia , Inflamação/microbiologia , Receptores do Fator de Necrose Tumoral/biossíntese , Fatores de Necrose Tumoral/biossíntese , Vagina/imunologia , Animais , Linhagem Celular , Colo do Útero/microbiologia , Citocina TWEAK , Feminino , Doenças dos Genitais Femininos/imunologia , Gonorreia/imunologia , Humanos , Inflamação/imunologia , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Lipopeptídeos/imunologia , Lipopeptídeos/farmacologia , Camundongos , Neisseria gonorrhoeae/imunologia , Receptor de TWEAK , Receptor 2 Toll-Like/imunologia , Vagina/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA