Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Infect Dis ; 9(12): 2401-2408, 2023 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-37955242

RESUMO

Exposure to environmental toxicants (such as dioxins) has been epidemiologically linked to adverse reproductive health outcomes, including placental inflammation and preterm birth. However, the molecular underpinnings that govern these outcomes in gravid reproductive tissues remain largely unclear. Placental macrophages (also known as Hofbauer cells) are crucial innate immune cells that defend the gravid reproductive tract and help promote maternal-fetal tolerance. We hypothesized that exposure to environmental toxicants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) could alter placental macrophage responses to inflammatory insults such as infection. To test this, placental macrophages were cultured in the presence or absence of TCDD and then infected with the perinatal pathogen Group B Streptococcus (GBS). Our results indicate that TCDD is lethal to placental macrophages at and above a 5 nM concentration and that sublethal dioxin exposure inhibits phagocytosis and cytokine production. Taken together, these results indicate that TCDD paralyzes placental macrophage responses to bacterial infection.


Assuntos
Dioxinas , Dibenzodioxinas Policloradas , Nascimento Prematuro , Humanos , Gravidez , Recém-Nascido , Feminino , Placenta , Dibenzodioxinas Policloradas/toxicidade , Macrófagos
2.
Am J Reprod Immunol ; 89(1): e13642, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36300889

RESUMO

Fetal or gestational membranes extend from the placenta to enclose the fetus and amniotic fluid. While the membranes spontaneously rupture at term in normal pregnancies, they can rupture prematurely before the onset of labor, termed preterm prelabor rupture of membranes (PPROM). PPROM can be triggered by bacterial infection or sterile inflammation in the membranes, known as chorioamnionitis (CAM). The membranes derive their tensile strength from a collagen-rich extracellular matrix (ECM); as such, understanding the enzymes and processes that can degrade the membrane ECM are of paramount importance. Matrix metalloproteinases (MMPs) are a class of enzymes capable of degrading collagen and other components of the ECM, and can be induced by inflammation. We used a scoping review to address the question of how MMP activity is associated with PPROM, particularly their induction due to sterile or nonsterile CAM. We have found that the most studied MMPs in PPROM were MMPs 2, 8, and 9. Additionally, some MMPs are constitutively active, while others are induced by inflammation. Mechanistic studies of the pathways that induce MMP activation are sparse, and this area is ripe for future studies. Targeting MMP activation could be a future strategy to delay or prevent PPROM.


Assuntos
Corioamnionite , Ruptura Prematura de Membranas Fetais , Trabalho de Parto , Feminino , Humanos , Recém-Nascido , Gravidez , Líquido Amniótico/metabolismo , Corioamnionite/metabolismo , Ruptura Prematura de Membranas Fetais/metabolismo , Trabalho de Parto/fisiologia , Metaloproteinases da Matriz/metabolismo
3.
Nat Commun ; 13(1): 5392, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36104331

RESUMO

Perinatal infection with Streptococcus agalactiae, or Group B Streptococcus (GBS), is associated with preterm birth, neonatal sepsis, and stillbirth. Here, we study the interactions of GBS with macrophages, essential sentinel immune cells that defend the gravid reproductive tract. Transcriptional analyses of GBS-macrophage co-cultures reveal enhanced expression of a gene encoding a putative metal resistance determinant, cadD. Deletion of cadD reduces GBS survival in macrophages, metal efflux, and resistance to metal toxicity. In a mouse model of ascending infection during pregnancy, the ΔcadD strain displays attenuated bacterial burden, inflammation, and cytokine production in gestational tissues. Furthermore, depletion of host macrophages alters cytokine expression and decreases GBS invasion in a cadD-dependent fashion. Our results indicate that GBS cadD plays an important role in metal detoxification, which promotes immune evasion and bacterial proliferation in the pregnant host.


Assuntos
Nascimento Prematuro , Streptococcus agalactiae , Animais , Citocinas , Feminino , Humanos , Recém-Nascido , Contagem de Leucócitos , Macrófagos/microbiologia , Metais , Camundongos , Gravidez , Nascimento Prematuro/microbiologia , Streptococcus agalactiae/genética
4.
J Matern Fetal Neonatal Med ; 35(14): 2795-2807, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32787482

RESUMO

The fetal membranes enclose the growing fetus and amniotic fluid. Preterm prelabor rupture of fetal membranes is a leading cause of preterm birth. Fetal membranes are composed of many different cell types, both structural and immune. These cells must coordinate functions for tensile strength and membrane integrity to contain the growing fetus and amniotic fluid. They must also balance immune responses to pathogens with maintaining maternal-fetal tolerance. Perturbation of this equilibrium can lead to preterm premature rupture of membranes without labor. In this review, we describe the formation of the fetal membranes to orient the reader, discuss some of the common forms of communication between the cell types of the fetal membranes, and delve into the methods used to tease apart this paracrine signaling within the membranes, including emerging technologies such as organ-on-chip models of membrane immunobiology.


Assuntos
Ruptura Prematura de Membranas Fetais , Nascimento Prematuro , Líquido Amniótico/metabolismo , Bioengenharia , Comunicação , Membranas Extraembrionárias/metabolismo , Feminino , Ruptura Prematura de Membranas Fetais/metabolismo , Humanos , Recém-Nascido , Nascimento Prematuro/metabolismo
5.
ACS Infect Dis ; 7(9): 2686-2696, 2021 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-34076405

RESUMO

Group B Streptococcus (GBS) is an encapsulated Gram-positive pathogen that causes ascending infections of the reproductive tract during pregnancy. The capsule of this organism is a critical virulence factor that has been implicated in a variety of cellular processes to promote pathogenesis. Primarily comprised of carbohydrates, the GBS capsule and its synthesis is driven by the capsule polysaccharide synthesis (cps) operon. The cpsE gene within this operon encodes a putative glycosyltransferase that is responsible for the transfer of a Glc-1-P from UDP-Glc to an undecaprenyl lipid molecule. We hypothesized that the cpsE gene product is important for GBS virulence and ascending infection during pregnancy. Our work demonstrates that a GBS cpsE mutant secretes fewer carbohydrates, has a reduced capsule, and forms less biofilm than the wild-type parental strain. We show that, compared to the parental strain, the ΔcpsE deletion mutant is more readily taken up by human placental macrophages and has a significantly attenuated ability to invade and proliferate in the mouse reproductive tract. Taken together, these results demonstrate that the cpsE gene product is an important virulence factor that aids in GBS colonization and invasion of the gravid reproductive tract.


Assuntos
Cápsulas Bacterianas , Placenta , Animais , Biofilmes , Feminino , Camundongos , Gravidez , Sorogrupo , Streptococcus agalactiae/genética
6.
Chembiochem ; 22(12): 2124-2133, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33755306

RESUMO

Group B Streptococcus (GBS) is an encapsulated Gram-positive human pathogen that causes invasive infections in pregnant hosts and neonates, as well as immunocompromised individuals. Colonization of the human host requires the ability to adhere to mucosal surfaces and circumnavigate the nutritional challenges and antimicrobial defenses associated with the innate immune response. Biofilm formation is a critical process to facilitate GBS survival and establishment of a replicative niche in the vertebrate host. Previous work has shown that the host responds to GBS infection by producing the innate antimicrobial glycoprotein lactoferrin, which has been implicated in repressing bacterial growth and biofilm formation. Additionally, lactoferrin is highly abundant in human breast milk and could serve a protective role against invasive microbial pathogens. This study demonstrates that human breast milk lactoferrin has antimicrobial and anti-biofilm activity against GBS and inhibits its adherence to human gestational membranes. Together, these results indicate that human milk lactoferrin could be used as a prebiotic chemotherapeutic strategy to limit the impact of bacterial adherence and biofilm formation on GBS-associated disease outcomes.


Assuntos
Antibacterianos/farmacologia , Lactoferrina/imunologia , Leite Humano/química , Streptococcus agalactiae/efeitos dos fármacos , Antibacterianos/química , Aderência Bacteriana/efeitos dos fármacos , Aderência Bacteriana/imunologia , Biofilmes/efeitos dos fármacos , Feminino , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Lactoferrina/química , Testes de Sensibilidade Microbiana , Streptococcus agalactiae/imunologia
7.
Clin Ther ; 43(2): 265-278, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33487441

RESUMO

PURPOSE: Maternal obesity, affecting nearly 1 in 4 pregnancies, is associated with increased circulating saturated fatty acids, such as palmitate. These fatty acids are implicated in placental inflammation, which may in turn exacerbate both maternal-fetal tolerance and responses to pathogens, such as group B Streptococcus. In this review, we address the question, "How do obesity and associated fatty acids influence placental inflammation?" METHODS: In this narrative review, we searched PubMed and Google Scholar using combinations of the key words placental inflammation or pregnancy and lipids, fatty acids, obesity, palmitate, or other closely related search terms. We also used references found within these articles that may have been absent from our original search queries. We analyzed methods and key results of these articles to compare and contrast their findings, which were occasionally at odds with each other. FINDINGS: Although obesity can be studied as a whole, complex phenomena with in vivo mouse models and human samples from patients with obesity, in vitro modeling often relies on the treatment of cells or tissues with ≥1 fatty acids and occasionally other compounds (eg, glucose and insulin). We found that palmitate, most commonly used in vitro to recreate hallmarks of obesity, induces apoptosis, oxidative stress, mitochondrial dysfunction, autophagy defects, and inflammasome activation in many placental cell types. We compare this to in vivo models of obesity wherever possible. We found that obesity as a whole may have more complex regulation of these phenomena (apoptosis, oxidative stress, mitochondrial dysfunction, autophagy defects, and inflammasome activation) compared with in vitro models of fatty acid treatment (primarily palmitate) because of the presence of unsaturated fatty acids (ie, oleate), which may have anti-inflammatory effects. IMPLICATIONS: The interaction of unsaturated fatty acids with saturated fatty acids may ameliorate many inflammatory effects of saturated fatty acids alone, which complicates interpretation of in vitro studies that focus on a particular fatty acid in isolation. This complication may explain why certain studies of obesity in vivo have differing outcomes from studies of specific fatty acids in vitro.


Assuntos
Ácidos Graxos/metabolismo , Inflamação/etiologia , Obesidade/metabolismo , Placenta/metabolismo , Complicações na Gravidez/metabolismo , Animais , Apoptose , Modelos Animais de Doenças , Feminino , Glucose/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Mitocôndrias/metabolismo , Palmitatos/metabolismo , Palmitatos/farmacologia , Placenta/patologia , Gravidez
8.
Am J Reprod Immunol ; 85(3): e13352, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32969101

RESUMO

PROBLEM: Gestational membrane (GM) infection provokes inflammation and can result in preterm prelabor rupture of membranes (PPROM). The choriodecidual layer of the GM includes decidual stromal cells (DSC), cytotrophoblasts (CTB), and macrophages (Mφ). Our laboratory has previously shown that DSCs suppress Mφ TNF-α production through secreted prostaglandin E2 . We hypothesized that CTBs would also inhibit Mφ cytokine expression through secreted mediators. METHOD OF STUDY: THP.1 Mφ-like cells with an NF-κB reporter construct or human blood monocyte-derived Mφ were co-cultured with the Jeg3 CTB cell line or primary human CTBs and challenged with group B streptococcus (GBS) or Toll-like receptor (TLR) agonists. Conditioned medium generated from CTB cultures was applied to Mφ cultures before infection or treatment. Alternatively, CTBs were co-incubated with, but physically separated from, Mφ and GBS or TLR-stimulated. NF-κB was assessed via alkaline phosphatase assay, and proinflammatory mediators were assessed by qRT-PCR and ELISA. RESULTS: CTBs suppressed GBS- or TLR-stimulated Mφ NF-κB activity, and TNF-α and MMP9 production. Direct physical contact between CTBs and Mφ was required for full immunosuppression. Immunosuppression could be overcome by increasing the ratio of Mφ to CTB. CONCLUSIONS: CTBs limit Mφ NF-κB activation and production of TNF-α and MMP9 through an as-yet unknown, cell-to-cell contact-mediated mechanism. This suppression is distinct from the PGE2 -mediated Mφ TNF-α suppression by DSC, suggesting that DSCs and CTBs regulate Mφ inflammation through distinct mechanisms. How Mφ integrates these signals in an intact GM will be paramount to determining causes and prevention of PPROM.


Assuntos
Âmnio/patologia , Decídua/patologia , Ruptura Prematura de Membranas Fetais/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus/fisiologia , Células Estromais/metabolismo , Adesão Celular , Feminino , Humanos , Tolerância Imunológica , Metaloproteinase 9 da Matriz/metabolismo , NF-kappa B/metabolismo , Gravidez , Transdução de Sinais , Células Estromais/patologia , Células THP-1 , Receptores Toll-Like/metabolismo , Trofoblastos , Fator de Necrose Tumoral alfa/metabolismo
9.
ACS Infect Dis ; 6(12): 3131-3140, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33170652

RESUMO

Streptococcus species are common causes of human infection. These Gram-positive, encapsulated bacterial pathogens infect diverse anatomic spaces, leading to infections including skin and soft tissue infection, endocarditis, pneumonia, meningitis, sinusitis, otitis media, chorioamnionitis, sepsis, and even death. Risk for streptococcal infection is highest in low- and middle-income countries where micronutrient deficiency is common. Epidemiological data reveal that vitamin D deficiency is associated with enhanced risk of streptococcal infection and cognate disease outcomes. Additionally, vitamin D improves antibacterial defenses by stimulating innate immune processes such as phagocytosis and enhancing production of reactive oxygen species (oxidative burst) and antimicrobial peptides (including cathelicidin and lactoferrin), which are important for efficient killing of bacteria. This review presents the most recent published work that studies interactions between the micronutrient vitamin D, the host immune system, and pathogenic streptococci as well as comparisons with other relevant infection models.


Assuntos
Anti-Infecciosos , Infecções Estreptocócicas , Deficiência de Vitamina D , Vitamina D , Feminino , Humanos , Imunidade , Gravidez , Streptococcus
10.
Placenta ; 90: 45-51, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-32056551

RESUMO

INTRODUCTION: There is an increasing prevalence of non-communicable diseases worldwide. Metabolic diseases such as obesity and gestational diabetes mellitus (GDM) increasingly affect women during pregnancy, which can harm pregnancy outcomes and the long-term health and wellbeing of exposed offspring. Both obesity and GDM have been associated with proinflammatory effects within the placenta, the critical organ governing fetal development. METHODS: The purpose of these studies was to model, in vitro, the effects of metabolic stress (high levels of glucose, insulin and saturated lipids) on placental macrophage biology, since these cells are the primary innate immune phagocyte within the placenta with roles in governing maternofetal immune tolerance and antimicrobial host defense. Macrophages were isolated from the villous core of term, human placentae delivered through nonlaboring, elective Cesarean sections and exposed to combinations of elevated glucose (30 mM), insulin (10 nM) and the saturated lipid palmitic acid (palmitate, 0.4 mM). RESULTS: We found that palmitate alone induced the activation of the nucleotide-binding oligomerization domain-like receptor (NLR) Family Pyrin Domain Containing 3 (NLRP3) inflammasome in placental macrophages, which was associated with increased interleukin 1 beta release and an increase in apoptotic cell death. Glucose and insulin neither provoked these effects nor augmented the impact of palmitate itself. DISCUSSION: Our findings confirm an impact of saturated fat on placental macrophage immune activation and could be relevant to the impact of metabolic stress in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Ácido Palmítico/farmacologia , Placenta/efeitos dos fármacos , Adulto , Citocinas/metabolismo , Feminino , Glucose/farmacologia , Humanos , Inflamassomos/metabolismo , Insulina/farmacologia , Macrófagos/metabolismo , Placenta/metabolismo , Gravidez
11.
Sci Adv ; 5(12): eaaw9051, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31840058

RESUMO

Epigenetic modifications play critical roles in inducing long-lasting immunological memory in innate immune cells, termed trained immunity. Whether similar epigenetic mechanisms regulate dendtritic cell (DC) function to orchestrate development of adaptive immunity remains unknown. We report that DCs matured with IFNγ and TNFα or matured in the lungs during invasive fungal infection with endogenous TNFα acquired a stable TNFα-dependent DC1 program, rendering them resistant to both antigen- and cytokine-induced alternative activation. TNFα-programmed DC1 had increased association of H3K4me3 with DC1 gene promoter regions. Furthermore, MLL1 inhibition blocked TNFα-mediated DC1 phenotype stabilization. During IFI, TNFα-programmed DC1s were required for the development of sustained TH1/TH17 protective immunity, and bone marrow pre-DCs exhibited TNFα-dependent preprogramming, supporting continuous generation of programmed DC1 throughout the infection. TNFα signaling, associated with epigenetic activation of DC1 genes particularly via H3K4me3, critically contributes to generation and sustenance of type 1/17 adaptive immunity and the immune protection against persistent infection.


Assuntos
Polaridade Celular , Citoproteção , Células Dendríticas/metabolismo , Epigênese Genética , Linfócitos T/citologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Polaridade Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Cryptococcus/efeitos dos fármacos , Cryptococcus/fisiologia , Citoproteção/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Feminino , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Imunomodulação/efeitos dos fármacos , Lisina/metabolismo , Metilação , Camundongos Endogâmicos CBA , Proteína de Leucina Linfoide-Mieloide/metabolismo , Fenótipo , Regiões Promotoras Genéticas/genética , Supressão Genética/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Fator de Necrose Tumoral alfa/farmacologia
13.
J Infect Dis ; 217(4): 656-666, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29253201

RESUMO

Background: Understanding the relationship between the levonorgestrel (LNG)-releasing intrauterine system (IUS) and sexually transmitted infections (STIs) is increasingly important as use of the LNG-IUS grows to include women at higher risk for STIs. This study assessed the impact of the LNG-IUS on development of Chlamydia trachomatis pelvic inflammatory disease, using a baboon model. Methods: Baboons with and those without the LNG-IUS were cervically inoculated with C. trachomatis and monitored daily, and cervical and fallopian tube swab specimens were collected weekly for C. trachomatis quantitation by nucleic acid amplification testing and culture. Vaginal swab specimens were collected for cytokine analysis, and serum samples were obtained for detection of C. trachomatis antibodies. Results: The LNG-IUS resulted in an increased C. trachomatis burden in the cervix, with the bacterial burden in the LNG-IUS group diverging from that in the non-LNG-IUS group by 6 weeks after infection. One of 7 baboons in the non-LNG-IUS group and 2 of 6 in the LNG-IUS group developed pelvic inflammatory disease, while 3 animals in each group met criteria suggestive of pelvic inflammatory disease. LNG-IUS increased baseline interleukin 8 levels but failed to further upregulate interleukin 8 during infection. In LNG-IUS recipients, early perturbations in the interleukin 1ß axis corresponded to decreased C. trachomatis clearance and increased T-helper type 2 immune responses. Conclusion: LNG-IUS use results in delayed clearance of C. trachomatis and might alter the reproductive tract immune environment.


Assuntos
Infecções por Chlamydia/patologia , Chlamydia trachomatis/isolamento & purificação , Anticoncepcionais Femininos/administração & dosagem , Dispositivos Intrauterinos/efeitos adversos , Levanogestrel/administração & dosagem , Doença Inflamatória Pélvica/patologia , Doenças Bacterianas Sexualmente Transmissíveis/patologia , Animais , Anticorpos Antibacterianos/sangue , Colo do Útero/microbiologia , Citocinas/análise , Modelos Animais de Doenças , Progressão da Doença , Tubas Uterinas/microbiologia , Feminino , Papio , Vagina/patologia
14.
J Immunol ; 198(9): 3548-3557, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28298522

RESUMO

The scavenger receptor macrophage receptor with collagenous structure (MARCO) promotes protective innate immunity against bacterial and parasitic infections; however, its role in host immunity against fungal pathogens, including the major human opportunistic fungal pathogen Cryptococcus neoformans, remains unknown. Using a mouse model of C. neoformans infection, we demonstrated that MARCO deficiency leads to impaired fungal control during the afferent phase of cryptococcal infection. Diminished fungal containment in MARCO-/- mice was accompanied by impaired recruitment of Ly6Chigh monocytes and monocyte-derived dendritic cells (moDC) and lower moDC costimulatory maturation. The reduced recruitment and activation of mononuclear phagocytes in MARCO-/- mice was linked to diminished early expression of IFN-γ along with profound suppression of CCL2 and CCL7 chemokines, providing evidence for roles of MARCO in activation of the CCR2 axis during C. neoformans infection. Lastly, we found that MARCO was involved in C. neoformans phagocytosis by resident pulmonary macrophages and DC. We conclude that MARCO facilitates early interactions between C. neoformans and lung-resident cells and promotes the production of CCR2 ligands. In turn, this contributes to a more robust recruitment and activation of moDC that opposes rapid fungal expansion during the afferent phase of cryptococcal infection.


Assuntos
Criptococose/imunologia , Cryptococcus neoformans/fisiologia , Células Dendríticas/imunologia , Pneumopatias Fúngicas/imunologia , Macrófagos/imunologia , Receptores Imunológicos/metabolismo , Receptores Depuradores/metabolismo , Animais , Células Cultivadas , Quimiocina CCL7/metabolismo , Células Dendríticas/microbiologia , Modelos Animais de Doenças , Humanos , Imunidade Inata , Interferon gama/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Receptores Imunológicos/genética , Receptores Depuradores/genética
15.
mBio ; 7(4)2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27406560

RESUMO

UNLABELLED: Anti-tumor necrosis factor alpha (anti-TNF-α) therapies have been increasingly used to treat inflammatory diseases and are associated with increased risk of invasive fungal infections, including Cryptococcus neoformans infection. Using a mouse model of cryptococcal infection, we investigated the mechanism by which disruption of early TNF-α signaling results in the development of nonprotective immunity against C. neoformans We found that transient depletion of TNF-α inhibited pulmonary fungal clearance and enhanced extrapulmonary dissemination of C. neoformans during the adaptive phase of the immune response. Higher fungal burdens in TNF-α-depleted mice were accompanied by markedly impaired Th1 and Th17 responses in the infected lungs. Furthermore, early TNF-α depletion also resulted in disrupted transcriptional initiation of the Th17 polarization program and subsequent upregulation of Th1 genes in CD4(+) T cells in the lung-associated lymph nodes (LALN) of C. neoformans-infected mice. These defects in LALN T cell responses were preceded by a dramatic shift from a classical toward an alternative activation of dendritic cells (DC) in the LALN of TNF-α-depleted mice. Taken together, our results indicate that early TNF-α signaling is required for optimal DC activation, and the initial Th17 response followed by Th1 transcriptional prepolarization of T cells in the LALN, which further drives the development of protective immunity against cryptococcal infection in the lungs. Thus, administration of anti-TNF-α may introduce a particularly greater risk for newly acquired fungal infections that require generation of protective Th1/Th17 responses for their containment and clearance. IMPORTANCE: Increased susceptibility to invasive fungal infections in patients on anti-TNF-α therapies underlines the need for understanding the cellular effects of TNF-α signaling in promoting protective immunity to fungal pathogens. Here, we demonstrate that early TNF-α signaling is required for classical activation and accumulation of DC in LALN of C. neoformans-infected mice. Subsequent transcriptional initiation of Th17 followed by Th1 programming in LALN results in pulmonary accumulation of gamma interferon- and interleukin-17A-producing T cells and effective fungal clearance. All of these crucial steps are severely impaired in mice that undergo anti-TNF-α treatment, consistent with their inability to clear C. neoformans This study identified critical interactions between cells of the innate immune system (DC), the emerging T cell responses, and cytokine networks with a central role for TNF-α which orchestrate the development of the immune protection against cryptococcal infection. This information will be important in aiding development and understanding the potential side effects of immunotherapies.


Assuntos
Criptococose/imunologia , Criptococose/prevenção & controle , Células Dendríticas/imunologia , Pneumopatias/imunologia , Pneumopatias/prevenção & controle , Transdução de Sinais , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Carga Bacteriana , Linfócitos T CD4-Positivos/imunologia , Cryptococcus neoformans/imunologia , Modelos Animais de Doenças , Pulmão/imunologia , Pulmão/microbiologia , Linfonodos/imunologia , Camundongos
16.
Future Microbiol ; 10(11): 1837-57, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26597428

RESUMO

This review discusses the unique contributions of dendritic cells (DCs) to T-cell priming and the generation of effective host defenses against Cryptococcus neoformans (C.neo) infection. We highlight DC subsets involved in the early and later stages of anticryptococcal immune responses, interactions between C.neo pathogen-associated molecular patterns and pattern recognition receptors expressed by DC, and the influence of DC on adaptive immunity. We emphasize recent studies in mouse models of cryptococcosis that illustrate the importance of DC-derived cytokines and costimulatory molecules and the potential role of DC epigenetic modifications that support maintenance of these signals throughout the immune response to C.neo. Lastly, we stipulate where these advances can be developed into new, immune-based therapeutics for treatment of this global pathogen.


Assuntos
Criptococose/microbiologia , Criptococose/patologia , Cryptococcus neoformans/imunologia , Células Dendríticas/imunologia , Interações Hospedeiro-Patógeno , Pneumopatias Fúngicas/microbiologia , Pneumopatias Fúngicas/patologia , Animais , Criptococose/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Pneumopatias Fúngicas/imunologia
17.
J Immunol ; 194(12): 5999-6010, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25972480

RESUMO

Numerous virulence factors expressed by Cryptococcus neoformans modulate host defenses by promoting nonprotective Th2-biased adaptive immune responses. Prior studies demonstrate that the heat shock protein 70 homolog, Ssa1, significantly contributes to serotype D C. neoformans virulence through the induction of laccase, a Th2-skewing and CNS tropic factor. In the present study, we sought to determine whether Ssa1 modulates host defenses in mice infected with a highly virulent serotype A strain of C. neoformans (H99). To investigate this, we assessed pulmonary fungal growth, CNS dissemination, and survival in mice infected with either H99, an SSA1-deleted H99 strain (Δssa1), and a complement strain with restored SSA1 expression (Δssa1::SSA1). Mice infected with the Δssa1 strain displayed substantial reductions in lung fungal burden during the innate phase (days 3 and 7) of the host response, whereas less pronounced reductions were observed during the adaptive phase (day 14) and mouse survival increased only by 5 d. Surprisingly, laccase activity assays revealed that Δssa1 was not laccase deficient, demonstrating that H99 does not require Ssa1 for laccase expression, which explains the CNS tropism we still observed in the Ssa1-deficient strain. Lastly, our immunophenotyping studies showed that Ssa1 directly promotes early M2 skewing of lung mononuclear phagocytes during the innate phase, but not the adaptive phase, of the immune response. We conclude that Ssa1's virulence mechanism in H99 is distinct and laccase-independent. Ssa1 directly interferes with early macrophage polarization, limiting innate control of C. neoformans, but ultimately has no effect on cryptococcal control by adaptive immunity.


Assuntos
Criptococose/imunologia , Criptococose/metabolismo , Cryptococcus neoformans/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Pneumopatias Fúngicas/imunologia , Pneumopatias Fúngicas/microbiologia , Macrófagos/imunologia , Imunidade Adaptativa , Animais , Encéfalo/metabolismo , Encéfalo/microbiologia , Encéfalo/patologia , Criptococose/mortalidade , Criptococose/patologia , Cryptococcus neoformans/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Regulação Fúngica da Expressão Gênica , Proteínas de Choque Térmico HSP70/genética , Imunidade Inata , Lacase/genética , Lacase/metabolismo , Leucócitos/imunologia , Leucócitos/patologia , Pneumopatias Fúngicas/mortalidade , Pneumopatias Fúngicas/patologia , Ativação de Macrófagos/imunologia , Camundongos , Mutação
18.
J Immunol ; 194(5): 2219-31, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25637026

RESUMO

Upon ingestion by macrophages, Cryptococcus neoformans can survive and replicate intracellularly unless the macrophages become classically activated. The mechanism enabling intracellular replication is not fully understood; neither are the mechanisms that allow classical activation to counteract replication. C. neoformans-induced lysosome damage was observed in infected murine bone marrow-derived macrophages, increased with time, and required yeast viability. To demonstrate lysosome damage in the infected host, we developed a novel flow cytometric method for measuring lysosome damage. Increased lysosome damage was found in C. neoformans-containing lung cells compared with C. neoformans-free cells. Among C. neoformans-containing myeloid cells, recently recruited cells displayed lower damage than resident cells, consistent with the protective role of recruited macrophages. The magnitude of lysosome damage correlated with increased C. neoformans replication. Experimental induction of lysosome damage increased C. neoformans replication. Activation of macrophages with IFN-γ abolished macrophage lysosome damage and enabled increased killing of C. neoformans. We conclude that induction of lysosome damage is an important C. neoformans survival strategy and that classical activation of host macrophages counters replication by preventing damage. Thus, therapeutic strategies that decrease lysosomal damage, or increase resistance to such damage, could be valuable in treating cryptococcal infections.


Assuntos
Criptococose/tratamento farmacológico , Cryptococcus neoformans/patogenicidade , Interferon gama/farmacologia , Pneumopatias Fúngicas/tratamento farmacológico , Lisossomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Animais , Criptococose/imunologia , Criptococose/microbiologia , Criptococose/patologia , Cryptococcus neoformans/imunologia , Interações Hospedeiro-Patógeno , Luz , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Pneumopatias Fúngicas/imunologia , Pneumopatias Fúngicas/microbiologia , Pneumopatias Fúngicas/patologia , Lisossomos/microbiologia , Lisossomos/patologia , Lisossomos/efeitos da radiação , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Processos Fotoquímicos , Cultura Primária de Células , Virulência
19.
J Neurochem ; 127(5): 592-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23952003

RESUMO

Microglia, the resident innate immune cells of the CNS, are the primary defenders against microbes and critical to CNS remodeling. Dysregulation of microglial behavior can lead to unchecked pro-inflammatory activity and subsequent neurodegeneration. The molecular mechanisms leading to chronic inflammation and microglial dysfunction in neurodegenerative diseases are not well-understood. It is known that patients with Presenilin 2 (PS2) mutations develop autosomal dominant Alzheimer disease. We have shown that a lack of normal PS2 function is associated with exaggerated microglia pro-inflammatory responses in vitro. To identify pathways by which PS2 regulates microglia and determine how PS2 dysfunction may lead to altered inflammatory pathways, we pursued an unbiased array approach to assess differential expression of microRNAs between murine PS2 knockout (KO) and wild-type microglia. We identified miR146, a negative regulator of monocyte pro-inflammatory response, as constitutively down-regulated in PS2 KO microglia. Consistent with a state of miR146 suppression, we found that PS2 KO microglia express higher levels of the miR146 target protein interleukin-1 receptor-associated kinase-1, and have increased NFκB transcriptional activity. We hypothesize that PS2 impacts microglial responses through modulation of miR146a. PS2 dysfunction, through aging or mutation, may contribute to neurodegeneration by influencing the pro-inflammatory behavior of microglia. Presenilin 2 (PS2), a membrane associated protease, has been implicated in the pathogenesis of Alzheimer disease. We have previously shown that PS2 plays an important role in curbing the proinflammatory response in microglia. Here, we report the novel finding that PS2 participates in maintaining the basal and cytokine induced expression of the innate immunity regulating microRNA, miR146. These data suggest one mechanism by which PS2 works to reign in proinflammatory microglial behavior and that PS2 dysfunction or deficiency could thus result in unchecked proinflammatory activation contributing to neurodegeneration.


Assuntos
MicroRNAs/metabolismo , Microglia/imunologia , Presenilina-2/genética , Animais , Animais Recém-Nascidos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/imunologia , Microglia/citologia , NF-kappa B/imunologia , NF-kappa B/metabolismo , Presenilina-2/imunologia , Cultura Primária de Células , Transcrição Gênica/imunologia
20.
Am J Respir Cell Mol Biol ; 45(3): 650-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21257927

RESUMO

Activation of the Fas/Fas ligand (FasL) system is associated with activation of apoptotic and proinflammatory pathways that lead to the development of acute lung injury. Previous studies in chimeric mice and macrophage-depleted mice suggested that the main effector cell in Fas-mediated lung injury is not a myeloid cell, but likely an epithelial cell. The goal of this study was to determine whether epithelial cells release proinflammatory cytokines after Fas activation, and to identify the relevant pathways. Incubation of the murine alveolar epithelial cell line, MLE-12, with the Fas-activating monoclonal antibody, Jo2, resulted in release of the CXC chemokine, KC, in a dose-dependent manner. KC release was not prevented by the pan-caspase inhibitor, zVAD.fmk. Silencing of the adaptor protein, MyD88, with small interfering (si)RNA resulted in attenuation of KC release in response to Jo2. Fas activation resulted in phosphorylation of the mitogen-activated kinases extracellular signal-regulated kinase (ERK) and c-Jun-N-terminal kinase (JNK), and pharmacologic inhibition of ERK and JNK attenuated KC release in a dose-response manner. Similarly, primary human small airways epithelial cells released IL-8 in response to soluble FasL, and this was abrogated by inhibition of JNK and ERK. In vivo confirmatory studies showed that MyD88-null mice are protected from Fas-induced acute lung injury. In summary, we conclude that Fas induces KC release in MLE-12 cells by a mechanism requiring MyD88, mitogen-activated protein kinases, and likely activator protein-1.


Assuntos
Quimiocina CXCL1/metabolismo , Células Epiteliais/citologia , Fator 88 de Diferenciação Mieloide/metabolismo , Alvéolos Pulmonares/citologia , Receptor fas/metabolismo , Animais , Relação Dose-Resposta a Droga , Proteína Ligante Fas/metabolismo , Interleucina-8/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , RNA Interferente Pequeno/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...