Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pathog Dis ; 73(9): ftv100, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26519447

RESUMO

Chlamydia trachomatis urogenital serovars are intracellular bacteria that parasitize human reproductive tract epithelium. As the principal cell type supporting bacterial replication, epithelial cells are central to Chlamydia immunobiology initially as sentries and innate defenders, and subsequently as collaborators in adaptive immunity-mediated bacterial clearance. In asymptomatic individuals who do not seek medical care a decisive struggle between C. trachomatis and host defenses occurs at the epithelial interface. For this study, we modeled the immunobiology of epithelial cells and macrophages lining healthy genital mucosa and inflamed/infected mucosa during the transition from innate to adaptive immunity. Upper reproductive tract epithelial cell line responses were compared to bone marrow-derived macrophages utilizing gene expression microarray technology. Those comparisons showed minor differences in the intrinsic innate defenses of macrophages and epithelial cells. Major lineage-specific differences in immunobiology relate to epithelial collaboration with adaptive immunity including an epithelial requirement for inflammatory cytokines to express MHC class II molecules, and a paucity and imbalance between costimulatory and coinhibitory ligands on epithelial cells that potentially limits sterilizing immunity (replication termination) to Chlamydia-specific T cells activated with limited or unconventional second signals.


Assuntos
Infecções por Chlamydia/microbiologia , Chlamydia muridarum/crescimento & desenvolvimento , Células Epiteliais/fisiologia , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Macrófagos/fisiologia , Infecções do Sistema Genital/microbiologia , Imunidade Adaptativa , Animais , Linhagem Celular , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/patologia , Células Epiteliais/microbiologia , Feminino , Imunidade Inata , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Análise em Microsséries , Modelos Biológicos , Mucosa/citologia , Mucosa/microbiologia , Infecções do Sistema Genital/imunologia , Infecções do Sistema Genital/patologia
2.
J Bacteriol ; 196(16): 2989-3001, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24914180

RESUMO

Chlamydiae are widespread Gram-negative pathogens of humans and animals. Salicylidene acylhydrazides, developed as inhibitors of type III secretion system (T3SS) in Yersinia spp., have an inhibitory effect on chlamydial infection. However, these inhibitors also have the capacity to chelate iron, and it is possible that their antichlamydial effects are caused by iron starvation. Therefore, we have explored the modification of salicylidene acylhydrazides with the goal to uncouple the antichlamydial effect from iron starvation. We discovered that benzylidene acylhydrazides, which cannot chelate iron, inhibit chlamydial growth. Biochemical and genetic analyses suggest that the derivative compounds inhibit chlamydiae through a T3SS-independent mechanism. Four single nucleotide polymorphisms were identified in a Chlamydia muridarum variant resistant to benzylidene acylhydrazides, but it may be necessary to segregate the mutations to differentiate their roles in the resistance phenotype. Benzylidene acylhydrazides are well tolerated by host cells and probiotic vaginal Lactobacillus species and are therefore of potential therapeutic value.


Assuntos
Antibacterianos/farmacologia , Compostos de Benzilideno/farmacologia , Chlamydia muridarum/efeitos dos fármacos , Chlamydia muridarum/crescimento & desenvolvimento , Análise Mutacional de DNA , Farmacorresistência Bacteriana , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA
3.
PLoS One ; 8(8): e69421, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967058

RESUMO

Macrophages can display a number of distinct phenotypes, known collectively as polarized macrophages. The best defined of these phenotypes are the classically-activated, interferon gamma (IFNγ)/LPS induced (M1) and alternatively-activated, IL-4 induced (M2) macrophages. The goal of this study is to characterize macrophage-Chlamydia interactions in the context of macrophage polarization. Here we use Chlamydia muridarum and murine bone-marrow derived macrophages to show Chlamydia does not induce M2 polarization in macrophages as a survival strategy. Unexpectedly, the infection of macrophages was silent with no upregulation of M1 macrophage-associated genes. We further demonstrate that macrophages polarized prior to infection have a differential capacity to control Chlamydia. M1 macrophages harbor up to 40-fold lower inclusion forming units (IFU) than non-polarized or M2 polarized macrophages. Gene expression analysis showed an increase in 16sRNA in M2 macrophages with no change in M1 macrophages. Suppressed Chlamydia growth in M1 macrophages correlated with the induction of a bacterial gene expression profile typical of persistence as evident by increased Euo expression and decreased Omp1 and Tal expression. Observations of permissive Chlamydia growth in non-polarized and M2 macrophages and persistence in M1 macrophages were supported through electron microscopy. This work supports the importance of IFNγ in the innate immune response to Chlamydia. However, demonstration that the M1 macrophages, despite an antimicrobial signature, fail to eliminate intracellular Chlamydia supports the notion that host-pathogen co-evolution has yielded a pathogen that can evade cellular defenses against this pathogen, and persist for prolonged periods of time in the host.


Assuntos
Polaridade Celular , Chlamydia muridarum/fisiologia , Espaço Intracelular/microbiologia , Macrófagos/citologia , Macrófagos/microbiologia , Viabilidade Microbiana , Animais , Chlamydia muridarum/crescimento & desenvolvimento , Macrófagos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos BALB C
4.
Infect Immun ; 79(10): 4019-28, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21807906

RESUMO

As arguably the most successful parasite, Chlamydia is an obligate intracellular bacterium replicating inside a vacuole of eukaryotic host cells. The chlamydial vacuole does not fuse with the defense cell organelle lysosome. We previously showed that chlamydial infection increases markers of autophagy, an innate antimicrobial activity requiring lysosomal function. However, the work presented here demonstrates that p62, an autophagy protein that is degraded in lysosomes, either remained unchanged or increased in chlamydia-infected human epithelial, mouse fibroblast, and mouse macrophage cell lines. In addition, the activities of three lysosomal enzymes analyzed were diminished in chlamydia-infected macrophages. Bafilomycin A1 (BafA), a specific inhibitor of vacuolar ATPase (vATPase) required for lysosomal function, increased the growth of the human pathogen Chlamydia trachomatis (L2) in wild-type murine fibroblasts and macrophages but inhibited growth in the autophagy-deficient ATG5(-/-) fibroblasts. BafA exhibited only slight inhibition or no effect on L2 growth in multiple human genital epithelial cell lines. In contrast to L2, the mouse pathogen Chlamydia muridarum (MoPn) was consistently inhibited by BafA in all cell lines examined, regardless of species origin and autophagy status. Finally, L2 but not MoPn grew more efficiently in the ATG5(-/-) cells than in wild-type cells. These results suggest that there are two types of vATPase-bearing organelles that regulate chlamydial infection: one supports chlamydial infection, while the other plays a defensive role through autophagy when cells are artificially infected with certain chlamydiae that have not been adapted to the host species.


Assuntos
Autofagia , Infecções por Chlamydia/patologia , Chlamydia muridarum/patogenicidade , Chlamydia trachomatis/patogenicidade , Organelas/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Linhagem Celular , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/microbiologia , Chlamydia muridarum/efeitos dos fármacos , Chlamydia muridarum/crescimento & desenvolvimento , Chlamydia trachomatis/efeitos dos fármacos , Chlamydia trachomatis/crescimento & desenvolvimento , Inibidores Enzimáticos/farmacologia , Células Epiteliais/microbiologia , Fibroblastos/microbiologia , Células HeLa , Humanos , Lisossomos/enzimologia , Lisossomos/metabolismo , Macrolídeos/farmacologia , Macrófagos/microbiologia , Camundongos , ATPases Vacuolares Próton-Translocadoras/antagonistas & inibidores
5.
J Immunol ; 186(6): 3615-21, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21296978

RESUMO

Mice that were intranasally vaccinated with live or dead Chlamydia muridarum with or without CpG-containing oligodeoxynucleotide 1862 elicited widely disparate levels of protective immunity to genital tract challenge. We found that the frequency of multifunctional T cells coexpressing IFN-γ and TNF-α with or without IL-2 induced by live C. muridarum most accurately correlated with the pattern of protection against C. muridarum genital tract infection, suggesting that IFN-γ(+)-producing CD4(+) T cells that highly coexpress TNF-α may be the optimal effector cells for protective immunity. We also used an immunoproteomic approach to analyze MHC class II-bound peptides eluted from dendritic cells (DCs) that were pulsed with live or dead C. muridarum elementary bodies (EBs). We found that DCs pulsed with live EBs presented 45 MHC class II C. muridarum peptides mapping to 13 proteins. In contrast, DCs pulsed with dead EBs presented only six MHC class II C. muridarum peptides mapping to three proteins. Only two epitopes were shared in common between the live and dead EB-pulsed groups. This study provides insights into the role of Ag presentation and cytokine secretion patterns of CD4(+) T effector cells that correlate with protective immunity elicited by live and dead C. muridarum. These insights should prove useful for improving vaccine design for Chlamydia trachomatis.


Assuntos
Apresentação de Antígeno/imunologia , Vacinas Bacterianas/imunologia , Infecções por Chlamydia/imunologia , Chlamydia muridarum/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Peptídeos/imunologia , Células Th1/imunologia , Doenças Vaginais/imunologia , Animais , Vacinas Bacterianas/administração & dosagem , Infecções por Chlamydia/prevenção & controle , Chlamydia muridarum/crescimento & desenvolvimento , Chlamydia muridarum/patogenicidade , Modelos Animais de Doenças , Feminino , Células HeLa , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunidade Celular , Corpos de Inclusão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Peptídeos/metabolismo , Células Th1/microbiologia , Doenças Vaginais/microbiologia , Doenças Vaginais/prevenção & controle
6.
J Immunol ; 185(11): 6911-20, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21037093

RESUMO

Chlamydia trachomatis serovars D-K are sexually transmitted intracellular bacterial pathogens that replicate in epithelial cells lining the human reproductive tract. It is clear from knockout mice and T cell depletion studies using Chlamydia muridarum that MHC class II and CD4 T cells are critical for clearing bacteria from the murine genital tract. It is not clear how CD4 T cells interact with infected epithelial cells to mediate bacterial clearance in vivo. Previous work using an epithelial tumor cell line showed that a Chlamydia-specific CD4 T cell clone was able to inhibit C. muridarum replication in vitro via induction of epithelial NO production. We have previously shown that Chlamydia-specific CD4 T cell clones can recognize and be activated by infected reproductive tract epithelial cells and block Chlamydia replication in them. We extend those observations by investigating the mechanism used by a panel of CD4 T cell clones to control Chlamydia replication in epithelial cells. We found that Chlamydia-specific CD4 T cell clones were cytolytic, but that cytolysis was not likely critical for controlling C. muridarum replication. For one, CD4 T cell clone-induced epithelial NO production was critical for controlling replication; however, the most potent CD4 T cell clones were dependent on T cell degranulation for replication control with only a minor additional contribution from NO production. We discuss our data as they relate to existing knockout mouse studies addressing mechanisms of T cell-mediated control of Chlamydia replication and their implications for intracellular epithelial pathogens in mouse models.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Chlamydia muridarum/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Óxido Nítrico/fisiologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/microbiologia , Chlamydia muridarum/crescimento & desenvolvimento , Células Clonais , Células Epiteliais/metabolismo , Feminino , Doenças dos Genitais Femininos/imunologia , Doenças dos Genitais Femininos/microbiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/biossíntese , Óxido Nítrico/farmacologia
7.
Nature ; 457(7230): 731-5, 2009 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19060882

RESUMO

The obligate intracellular bacterium Chlamydia trachomatis survives and replicates within a membrane-bound vacuole, termed the inclusion, which intercepts host exocytic pathways to obtain nutrients. Like many other intracellular pathogens, C. trachomatis has a marked requirement for host cell lipids, such as sphingolipids and cholesterol, produced in the endoplasmic reticulum and the Golgi apparatus. However, the mechanisms by which intracellular pathogens acquire host cell lipids are not well understood. In particular, no host cell protein responsible for transporting Golgi-derived lipids to the chlamydial inclusions has yet been identified. Here we show that Chlamydia infection in human epithelial cells induces Golgi fragmentation to generate Golgi ministacks surrounding the bacterial inclusion. Ministack formation is triggered by the proteolytic cleavage of the Golgi matrix protein golgin-84. Inhibition of golgin-84 truncation prevents Golgi fragmentation, causing a block in lipid acquisition and maturation of C. trachomatis. Golgi fragmentation by means of RNA-interference-mediated knockdown of distinct Golgi matrix proteins before infection enhances bacterial maturation. Our data functionally connect bacteria-induced golgin-84 cleavage, Golgi ministack formation, lipid acquisition and intracellular pathogen growth. We show that C. trachomatis subverts the structure and function of an entire host cell organelle for its own advantage.


Assuntos
Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/patogenicidade , Complexo de Golgi/microbiologia , Complexo de Golgi/patologia , Chlamydia muridarum/crescimento & desenvolvimento , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Técnicas de Silenciamento de Genes , Complexo de Golgi/metabolismo , Proteínas da Matriz do Complexo de Golgi , Células HeLa , Humanos , Metabolismo dos Lipídeos , Proteínas de Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Interferência de RNA , Proteínas de Transporte Vesicular
8.
Mol Hum Reprod ; 13(12): 863-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17954522

RESUMO

Surfactant protein D (SP-D) plays a role in innate immunity in the lung and is expressed at many other mucosal surfaces throughout the human body. In this study, we show that SP-D mRNA and protein are present in the murine female reproductive tract; i.e. in the vagina, cervix, uterus and oviduct. SP-D protein is primarily localized to epithelial cells lining the genital tract and is also present in secretory material within the lumen of the uterus and cervix. The levels of SP-D mRNA in the uterus vary by a factor of 10 during the estrous cycle with peak levels present at estrus and the lowest levels at diestrus. In contrast, SP-D mRNA levels in the lung do not change during the estrous cycle. Since SP-D is an innate host defense protein present in the mouse reproductive tract, we studied the influence of infection on SP-D levels in vivo. We found that Chlamydia muridarum infection caused an increase in the SP-D protein content of reproductive tract epithelial cells. These data are suggestive that SP-D may play a role in innate immunity in the female reproductive tract in vivo.


Assuntos
Regulação da Expressão Gênica , Genitália Feminina/metabolismo , Proteína D Associada a Surfactante Pulmonar/genética , Animais , Colo do Útero/metabolismo , Colo do Útero/microbiologia , Infecções por Chlamydia/genética , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/microbiologia , Chlamydia muridarum/crescimento & desenvolvimento , Células Epiteliais/metabolismo , Feminino , Genitália Feminina/imunologia , Genitália Feminina/microbiologia , Imunidade Inata , Immunoblotting , Pulmão/metabolismo , Camundongos , Oviductos/metabolismo , Oviductos/microbiologia , Proteína D Associada a Surfactante Pulmonar/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Útero/metabolismo , Útero/microbiologia , Vagina/metabolismo , Vagina/microbiologia
9.
J Immunol ; 179(6): 3707-14, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17785807

RESUMO

Ligation of the purinergic receptor, P2X7R, with its agonist ATP has been previously shown to inhibit intracellular infection by chlamydiae and mycobacteria in macrophages. The effect of P2X7R on chlamydial infection had never been investigated in the preferred target cells of chlamydiae, cervical epithelial cells, nor in vaginally infected mice. In this study, we show that treatment of epithelial cells with P2X7R agonists inhibits partially Chlamydia infection in epithelial cells. Chelation of ATP with magnesium or pretreatment with a P2X7R antagonist blocks the inhibitory effects of ATP. Similarly to previous results obtained with macrophages, ATP-mediated inhibition of infection in epithelial cells requires activation of host-cell phospholipase D. Vaginal infection was also more efficient in P2X7R-deficient mice, which also displayed a higher level of acute inflammation in the endocervix, oviduct, and mesosalpingeal tissues than in infected wild-type mice. However, secretion of IL-1beta, which requires P2X7R ligation during infection by other pathogens, was decreased mildly and only at short times of infection. Taken together, these results suggest that P2X7R affects Chlamydia infection by directly inhibiting infection in epithelial cells, rather than through the ability of P2X7R to modulate IL-1beta secretion.


Assuntos
Colo do Útero/metabolismo , Infecções por Chlamydia/patologia , Infecções por Chlamydia/prevenção & controle , Doenças dos Genitais Femininos/prevenção & controle , Receptores Purinérgicos P2/fisiologia , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/fisiologia , Animais , Linhagem Celular Tumoral , Colo do Útero/imunologia , Colo do Útero/microbiologia , Infecções por Chlamydia/genética , Infecções por Chlamydia/imunologia , Chlamydia muridarum/crescimento & desenvolvimento , Chlamydia muridarum/imunologia , Doença Crônica , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Tubas Uterinas/metabolismo , Tubas Uterinas/patologia , Feminino , Doenças dos Genitais Femininos/imunologia , Doenças dos Genitais Femininos/metabolismo , Doenças dos Genitais Femininos/microbiologia , Doenças dos Genitais Femininos/patologia , Células HeLa , Humanos , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Agonistas do Receptor Purinérgico P2 , RNA Mensageiro/biossíntese , Receptores Purinérgicos P2/deficiência , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7
10.
Infect Immun ; 74(1): 225-38, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368976

RESUMO

Gamma interferon (IFN-gamma)-induced effector mechanisms have potent antichlamydial activities that are critical to host defense. The most prominent and well-studied effectors are indoleamine dioxygenase (IDO) and nitric oxide (NO) synthase. The relative contributions of these mechanisms as inhibitors of chlamydial in vitro growth have been extensively studied using different host cells, induction mechanisms, and chlamydial strains with conflicting results. Here, we have undertaken a comparative analysis of cytokine- and lipopolysaccharide (LPS)-induced IDO and NO using an extensive assortment of human and murine host cells infected with human and murine chlamydial strains. Following cytokine (IFN-gamma or tumor necrosis factor alpha) and/or LPS treatment, the majority of human cell lines induced IDO but failed to produce NO. Conversely, the majority of mouse cell lines studied produced NO, not IDO. Induction of IDO in human cell lines inhibited growth of L2 and mouse pneumonitis agent, now referred to as Chlamydia muridarum MoPn equally in all but two lines, and inhibition was completely reversible by the addition of tryptophan. IFN-gamma treatment of mouse cell lines resulted in substantially greater reduction of L2 than MoPn growth. However, despite elevated NO production by murine cells, blockage of NO synthesis with the l-arginine analogue N-monomethyl-l-arginine only partially rescued chlamydial growth, suggesting the presence of another IFN-gamma-inducible antichlamydial mechanism unique to murine cells. Moreover, NO generated from the chemical nitric oxide donor sodium nitroprusside showed little direct effect on chlamydial infectivity or growth, indicating a natural resistance to NO. Finally, IFN-gamma-inducible IDO expression in human HeLa cells was inhibited following exogenous NO treatment, resulting in a permissive environment for chlamydial growth. In summary, cytokine- and LPS-inducible effectors produced by human and mouse cells differ and, importantly, these host-specific effector responses result in chlamydial strain-specific antimicrobial activities.


Assuntos
Chlamydia muridarum/crescimento & desenvolvimento , Chlamydia muridarum/imunologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/imunologia , Interferon gama/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Infecções por Chlamydia/enzimologia , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/prevenção & controle , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Interferon gama/genética , Lipopolissacarídeos/farmacologia , Camundongos , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo II/fisiologia
11.
Microbes Infect ; 8(2): 478-86, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16297651

RESUMO

Chlamydiae alter apoptosis of host target cells, which regulates their growth. Cyclooxygenase-2 (COX-2), the rate-limiting enzyme for prostaglandin E2 (PGE2) production, modulates epithelial cell survival. We addressed whether endogenous PGE2 alters chlamydial growth or apoptosis of epithelial cells infected with Chlamydia muridarum. PGE2 is secreted by infected host cells in the genital tract (GT). Using immunohistochemical techniques, we found that COX-2 enzyme was localized to epithelial cells in the GT in vivo. Pellets of the COX-2 enzyme inhibitor, NS-398, and placebo were implanted in mice subcutaneously and released a constant amount of these chemicals throughout the infection. NS-398-treated mice were found to exhibit 10-fold lower bacterial load than the placebo group on day 3 post infection, suggesting disruption of the chlamydial developmental cycle. To prove this, the human lung adenocarcinoma cell line A549 was then infected with different MOIs of C. muridarum in the presence of multiple concentrations of NS-398 in vitro. There was no difference in inclusion forming units (IFUs) between NS-389-treated and untreated cells. We also found no alterations in C. muridarum IFUs in A549 cells transfected with a 2.0 kb cDNA fragment of human COX-2 cloned in the sense (S) or anti-sense (AS) orientation. However, the inclusion size was reduced and the number of EB was significantly diminished during reinfection in AS-transfected cells. In addition, the absence of COX-2 did not significantly modify apoptosis in infected cells. In total, COX-2 deficiency reduces the infectious burden in vivo and may modulate transmission of the organism.


Assuntos
Chlamydia muridarum/patogenicidade , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/microbiologia , Animais , Apoptose , Linhagem Celular Tumoral , Infecções por Chlamydia/microbiologia , Chlamydia muridarum/crescimento & desenvolvimento , Ciclo-Oxigenase 2/genética , Dinoprostona/metabolismo , Feminino , Doenças dos Genitais Femininos/microbiologia , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vagina/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA