Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Allergy Clin Immunol ; 142(1): 120-129.e6, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28916188

RESUMEN

BACKGROUND: Group B Streptococcus (GBS) or Streptococcus agalactiae are ß-hemolytic gram-positive bacteria that colonize the lower genital tracts of women and are frequently associated with infections during pregnancy. Innate immune defenses are critical for controlling GBS dissemination and systemic infection. Mast cells are resident sentinel cells that come into contact with pathogens early during colonization and infection. OBJECTIVE: We aimed to investigate the contribution of chymase to systemic GBS infection and rates of preterm birth. METHODS: Pharmacologic and genetic approaches using mice deficient in mast cell protease (MCPT) 4, the mouse functional homologue of human chymase, were used. RESULTS: Our studies show that mast cells release a protease with chymotrypsin-like cleavage specificity in response to GBS. Additionally, increased GBS systemic infection and preterm births were observed in MCPT4-deficient mice versus MCPT4-sufficient mice. Furthermore, we observed that proteolytic cleavage of the host extracellular matrix protein fibronectin by peritoneal cell-derived mast cell lysates diminished GBS adherence. Consistent with this observation, the increase in GBS dissemination and preterm births observed in MCPT4-deficient mice was abolished when GBS was deficient in expression of the fibronectin-binding protein SfbA. CONCLUSIONS: Taken together, our results suggest that the protective effect of MCPT4 against GBS dissemination and preterm labor can be attributed in part to MCPT4-mediated proteolysis of fibronectin. Our studies reveal a novel role of mast cells in defense against bacterial infections.


Asunto(s)
Mastocitos/inmunología , Serina Endopeptidasas/inmunología , Infecciones Estreptocócicas/inmunología , Animales , Quimasas/inmunología , Femenino , Mastocitos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Complicaciones Infecciosas del Embarazo/enzimología , Complicaciones Infecciosas del Embarazo/inmunología , Nacimiento Prematuro/inmunología , Nacimiento Prematuro/microbiología
2.
Biol Reprod ; 98(3): 396-407, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29155939

RESUMEN

Streptococcus agalactiae (group B streptococcus [GBS]) infection in pregnant women is the leading cause of infectious neonatal morbidity and mortality in the United States. Although inflammation during infection has been associated with preterm birth, the contribution of GBS to preterm birth is less certain. Moreover, the early mechanisms by which GBS interacts with the gestational tissue to affect adverse pregnancy outcomes are poorly understood. We hypothesized that short-term GBS inoculation activates pathways related to inflammation and premature birth in human extraplacental membranes. We tested this hypothesis using GBS-inoculated human extraplacental membranes in vitro. In agreement with our hypothesis, a microarray-based transcriptomics analysis of gene expression changes in GBS-inoculated membranes revealed that GBS activated pathways related to inflammation and preterm birth with significant gene expression changes occurring as early as 4 h postinoculation. In addition, pathways related to DNA replication and repair were downregulated with GBS treatment. Conclusions based on our transcriptomics data were further supported by responses of prostaglandin E2 (PGE2), and matrix metalloproteinases 1 (MMP1) and 3 (MMP3), all of which are known to be involved in parturition and premature rupture of membranes. These results support our initial hypothesis and provide new information on molecular targets of GBS infection in human extraplacental membranes.


Asunto(s)
Membranas Extraembrionarias/metabolismo , Rotura Prematura de Membranas Fetales/metabolismo , Nacimiento Prematuro/metabolismo , Infecciones Estreptocócicas/metabolismo , Streptococcus agalactiae , Transcriptoma , Dinoprostona/metabolismo , Membranas Extraembrionarias/microbiología , Femenino , Rotura Prematura de Membranas Fetales/microbiología , Humanos , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Embarazo , Nacimiento Prematuro/microbiología , Infecciones Estreptocócicas/microbiología
3.
Am J Obstet Gynecol ; 218(4): 438.e1-438.e16, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29475580

RESUMEN

BACKGROUND: Most early preterm births are associated with intraamniotic infection and inflammation, which can lead to systemic inflammation in the fetus. The fetal inflammatory response syndrome describes elevations in the fetal interleukin-6 level, which is a marker for inflammation and fetal organ injury. An understanding of the effects of inflammation on fetal cardiac development may lead to insight into the fetal origins of adult cardiovascular disease. OBJECTIVE: The purpose of this study was to determine whether the fetal inflammatory response syndrome is associated with disruptions in gene networks that program fetal cardiac development. STUDY DESIGN: We obtained fetal cardiac tissue after necropsy from a well-described pregnant nonhuman primate model (pigtail macaque, Macaca nemestrina) of intrauterine infection (n=5) and controls (n=5). Cases with the fetal inflammatory response syndrome (fetal plasma interleukin-6 >11 pg/mL) were induced by either choriodecidual inoculation of a hypervirulent group B streptococcus strain (n=4) or intraamniotic inoculation of Escherichia coli (n=1). RNA and protein were extracted from fetal hearts and profiled by microarray and Luminex (Millipore, Billerica, MA) for cytokine analysis, respectively. Results were validated by quantitative reverse transcriptase polymerase chain reaction. Statistical and bioinformatics analyses included single gene analysis, gene set analysis, Ingenuity Pathway Analysis (Qiagen, Valencia, CA), and Wilcoxon rank sum. RESULTS: Severe fetal inflammation developed in the context of intraamniotic infection and a disseminated bacterial infection in the fetus. Interleukin-6 and -8 in fetal cardiac tissues were elevated significantly in fetal inflammatory response syndrome cases vs controls (P<.05). A total of 609 probe sets were expressed differentially (>1.5-fold change, P<.05) in the fetal heart (analysis of variance). Altered expression of select genes was validated by quantitative reverse transcriptase polymerase chain reaction that included several with known functions in cardiac injury, morphogenesis, angiogenesis, and tissue remodeling (eg, angiotensin I converting enzyme 2, STEAP family member 4, natriuretic peptide A, and secreted frizzled-related protein 4; all P<.05). Multiple gene sets and pathways that are involved in cardiac morphogenesis and vasculogenesis were downregulated significantly by gene set and Ingenuity Pathway Analysis (hallmark transforming growth factor beta signaling, cellular morphogenesis during differentiation, morphology of cardiovascular system; all P<.05). CONCLUSION: Disruption of gene networks for cardiac morphogenesis and vasculogenesis occurred in the preterm fetal heart of nonhuman primates with preterm labor, intraamniotic infection, and severe fetal inflammation. Inflammatory injury to the fetal heart in utero may contribute to the development of heart disease later in life. Development of preterm labor therapeutics must also target fetal inflammation to lessen organ injury and potential long-term effects on cardiac function.


Asunto(s)
Enfermedades Fetales/metabolismo , Miocardio/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Factor Natriurético Atrial/genética , Biomarcadores/metabolismo , Corioamnionitis/metabolismo , Regulación hacia Abajo , Femenino , Corazón/microbiología , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Macaca nemestrina , Proteínas de la Membrana/genética , Análisis por Micromatrices , Modelos Animales , Trabajo de Parto Prematuro , Oxidorreductasas/genética , Peptidil-Dipeptidasa A/genética , Embarazo , Proteínas Proto-Oncogénicas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba
4.
Infect Immun ; 83(10): 3909-17, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26195546

RESUMEN

The mechanisms underlying fetal lung injury remain poorly defined. MicroRNAs (miRNAs) are small noncoding, endogenous RNAs that regulate gene expression and have been implicated in the pathogenesis of lung disease. Using a nonhuman primate model of choriodecidual infection, we sought to determine if differentially expressed miRNAs were associated with acute fetal lung injury. After inoculating 10 chronically catheterized pregnant monkeys (Macaca nemestrina) with either group B streptococcus (GBS) at 1 × 10(6) CFU (n = 5) or saline (n = 5) in the choriodecidual space, we extracted fetal lung mRNA and miRNA and profiled the changes in expression by microarray analysis. We identified 9 differentially expressed miRNAs in GBS-exposed fetal lungs, but of these, only miR-155-5p was validated by quantitative reverse transcription-PCR (P = 0.02). Significantly elevated miR-155-5p expression was also observed when immortalized human fetal airway epithelial (FeAE) cells were exposed to proinflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor alpha [TNF-α]). Overexpression of miR-155-5p in FeAE cells in turn increased the production of IL-6 and CXCL10/gamma interferon-induced protein 10, which are implicated in leukocyte recruitment but also in protection from lung injury. Interestingly, while miR-155-5p decreased fibroblast growth factor 9 (FGF9) expression in a luciferase reporter assay, FGF9 levels were actually increased in GBS-exposed fetal lungs in vivo. FGF9 overexpression is associated with abnormal lung development. Thus, upregulation of miR-155-5p may serve as a compensatory mechanism to lessen the increase in FGF9 and prevent aberrant lung development. Understanding the complicated networks regulating lung development in the setting of infection is a key step in identifying how to prevent fetal lung injury leading to bronchopulmonary dysplasia.


Asunto(s)
Enfermedades Fetales/genética , Enfermedades Fetales/microbiología , Pulmón/metabolismo , Infecciones Estreptocócicas/embriología , Infecciones Estreptocócicas/genética , Streptococcus/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Enfermedades Fetales/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/crecimiento & desarrollo , Pulmón/microbiología , Macaca nemestrina , Masculino , Embarazo , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Infect Immun ; 82(11): 4587-95, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25135682

RESUMEN

Group B Streptococcus (GBS) causes severe disease in neonates, the elderly, and immunocompromised individuals. GBS species are highly diverse and can be classified by serotype and multilocus sequence typing. Sequence type 17 (ST-17) strains cause invasive neonatal disease more frequently than strains of other STs. Attachment and invasion of host cells are key steps in GBS pathogenesis. We investigated whether four serotype III strains representing ST-17 (two strains), ST-19, and ST-23 differ in their abilities to attach to and invade both decidual cells and lung epithelial cells. Virulence gene expression following host cell association and exposure to amnion cells was also tested. The ST-17 strains differed in their abilities to attach to and invade decidual cells, whereas there were no differences with lung epithelial cells. The ST-19 and ST-23 strains, however, attached to and invaded decidual cells less than both ST-17 strains. Although the ST-23 strain attached to lung epithelial cells better than ST-17 and -19 strains, none of the strains effectively invaded the lung epithelial cells. Notably, the association with host cells resulted in the differential expression of several virulence genes relative to basal expression levels. Similar expression patterns of some genes were observed regardless of cell type used. Collectively, these results show that GBS strains differ in their abilities to attach to distinct host cell types and express key virulence genes that are relevant to the disease process. Enhancing our understanding of pathogenic mechanisms could aid in the identification of novel therapeutic targets or vaccine candidates that could potentially decrease morbidity and mortality associated with neonatal infections.


Asunto(s)
Decidua/citología , Células Epiteliales/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Pulmón/citología , Streptococcus agalactiae/metabolismo , Streptococcus agalactiae/patogenicidad , Línea Celular , Femenino , Humanos , Streptococcus agalactiae/clasificación , Streptococcus agalactiae/genética , Virulencia
6.
Am J Reprod Immunol ; 91(1): e13807, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38282602

RESUMEN

BACKGROUNDS: Infection during pregnancy is a significant public health concern due to the increased risk of adverse birth outcomes. Group B Streptococcus or Streptococcus agalactiae (GBS) stands out as a major bacterial cause of neonatal morbidity and mortality. We aimed to explore the involvement of reactive oxygen species (ROS) and oxidative stress pathways in pro-inflammatory responses within human fetal membrane tissue, the target tissue of acute bacterial chorioamnionitis. METHODS: We reanalyzed transcriptomic data from fetal membrane explants inoculated with GBS to assess the impact of GBS on oxidative stress and ROS genes/pathways. We conducted pathway enrichment analysis of transcriptomic data using the Database for Annotation, Visualization and Integrated Discovery (DAVID), a web-based functional annotation/pathway enrichment tool. Subsequently, we conducted ex vivo experiments to test the hypothesis that antioxidant treatment could inhibit pathogen-stimulated inflammatory responses in fetal membranes. RESULTS: Using DAVID analysis, we found significant enrichment of pathways related to oxidative stress or ROS in GBS-inoculated human fetal membranes, for example, "Response to Oxidative Stress" (FDR = 0.02) and "Positive Regulation of Reactive Oxygen Species Metabolic Process" (FDR = 2.6*10-4 ). There were 31 significantly changed genes associated with these pathways, most of which were upregulated after GBS inoculation. In ex vivo experiments with choriodecidual membrane explants, our study showed that co-treatment with N-acetylcysteine (NAC) effectively suppressed the release of pro-inflammatory cytokines (IL-6, IL-8, TNF-α) and prostaglandin PGE2, compared to GBS-treated explants (p < .05 compared to GBS-treated samples without NAC co-treatment). Furthermore, NAC treatment inhibited the release of cytokines and PGE2 stimulated by lipoteichoic acid (LTA) and lipopolysaccharide (LPS) in whole membrane explants (p < .05 compared to LTA or LPS-treated samples without NAC co-treatment). CONCLUSIONS: Our study sheds light on the potential roles of ROS in governing the innate immune response to GBS infection, offering insights for developing strategies to mitigate GBS-related adverse outcomes.


Asunto(s)
Corioamnionitis , Infecciones Estreptocócicas , Ácidos Teicoicos , Embarazo , Femenino , Recién Nacido , Humanos , Citocinas/metabolismo , Lipopolisacáridos/farmacología , Antioxidantes/farmacología , Antioxidantes/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Acetilcisteína/metabolismo , Dinoprostona/metabolismo , Prostaglandinas/metabolismo , Streptococcus agalactiae , Membranas Extraembrionarias/metabolismo
7.
Toxicology ; 483: 153371, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36396003

RESUMEN

Numerous Superfund sites are contaminated with the volatile organic chemical trichloroethylene (TCE). In women, exposure to TCE in pregnancy is associated with reduced birth weight. Our previous study reported that TCE exposure in pregnant rats decreased fetal weight and elevated oxidative stress biomarkers in placentae, suggesting placental injury as a potential mechanism of TCE-induced adverse birth outcomes. In this study, we investigated if co-exposure with the antioxidant N-acetylcysteine (NAC) attenuates TCE exposure effects on RNA expression. Timed-pregnant Wistar rats were exposed orally to 480 mg TCE/kg/day on gestation days 6-16. Exposure of 200 mg NAC/kg/day alone or as a pre/co-exposure with TCE occurred on gestation days 5-16 to stimulate antioxidant genes prior to TCE exposure. Tissue was collected on gestation day 16. In male and female placentae, we evaluated TCE- and/or NAC-induced changes to gene expression and pathway enrichment analyses using false discovery rate (FDR) and fold-change criteria. In female placentae, exposure to TCE caused significant differential expression 129 genes while the TCE+NAC altered 125 genes, compared with controls (FDR< 0.05 + fold-change >1). In contrast, in male placentae TCE exposure differentially expressed 9 genes and TCE+NAC differentially expressed 35 genes, compared with controls (FDR< 0.05 + fold-change >1). NAC alone did not significantly alter gene expression in either sex. Differentially expressed genes observed with TCE exposure were enriched in mitochondrial biogenesis and oxidative phosphorylation pathways in females whereas immune system pathways and endoplasmic reticulum stress pathways were differentially expressed in both sexes (FDR<0.05). TCE treatment was differentially enriched for genes regulated by the transcription factors ATF6 (both sexes) and ATF4 (males only), indicating a cellular condition triggered by misfolded proteins during endoplasmic reticulum stress. This study demonstrates novel genes and pathways involved in TCE-induced placental injury and showed antioxidant co-treatment largely did not attenuate TCE exposure effects.


Asunto(s)
Tricloroetileno , Femenino , Masculino , Ratas , Embarazo , Animales , Tricloroetileno/toxicidad , Tricloroetileno/metabolismo , Acetilcisteína/farmacología , Ratas Wistar , Antioxidantes/farmacología , Placenta/metabolismo
8.
Toxicol In Vitro ; 84: 105429, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35811015

RESUMEN

Studies have shown that the trichloroethylene metabolite S-(1,2-dichlorovinyl)-l-cysteine (DCVC) inhibits cytokine secretion in pathogen stimulated fetal membrane tissue but little is known about the mechanism for these effects, including which cell types or transcriptomic pathways are impacted. Macrophages play a critical role in fetal membrane immune responses during infection. We tested the hypothesis that DCVC inhibits lipopolysaccharide (LPS) stimulated inflammation pathways in macrophage-like THP-1 cells. We treated THP-1 cells for 24 h then treated with 1, 5, or 10 µM DCVC for 24 h. After a 4 h incubation with lipopolysaccharide (LPS), we collected RNA and cell media. We performed transcriptomic analysis using RNA sequencing for 5 µM DCVC treatments and quantified cytokine release (IL-1ß, IL-6, and TNF-α) for 1, 5 and 10 µM DCVC treatments. RNA sequencing analysis revealed 1399 differentially expressed genes (FDR < 0.05 and log 2 fold change magnitude>2.5) in cells co-treated with DCVC and LPS compared to LPS alone. For example, TNF had a log2(fold-change) = -3.5 with the addition of DCVC. Pathways downregulated (adjusted p-value<0.05) in DCVC+LPS treatments versus LPS-only treatments included: "acute inflammatory response", "production of molecular mediator of immune response" and "phagocytosis". LPS increased IL-1ß, IL-6, and TNF-α levels in culture media (p < 0.001), but this was inhibited by co-treatment with DCVC (p < 0.001 for LPS vs. LPS + DCVC treatments). Our results demonstrate that DCVC suppresses inflammatory responses in macrophages.


Asunto(s)
Cisteína , Tricloroetileno , Humanos , Inflamación/inducido químicamente , Interleucina-6 , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Transcriptoma , Tricloroetileno/metabolismo , Tricloroetileno/toxicidad , Factor de Necrosis Tumoral alfa
9.
Front Physiol ; 11: 565, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32547423

RESUMEN

In addition to providing a physical compartment for gestation, the fetal membranes (FM) are an active immunological barrier that provides defense against pathogenic microorganisms that ascend the gravid reproductive tract. Pathogenic infection of the gestational tissues (FM and placenta) is a leading known cause of preterm birth (PTB). Some environmental toxicants decrease the capacity for organisms to mount an immune defense against pathogens. For example, the immunosuppressive effects of the widespread environmental contaminant trichloroethylene (TCE) are documented for lung infection with Streptococcus zooepidemicus. Group B Streptococcus (GBS; Streptococcus agalactiae) is a bacterial pathogen that is frequently found in the female reproductive tract and can colonize the FM in pregnant women. Work in our laboratory has demonstrated that a bioactive TCE metabolite, S-(1, 2-dichlorovinyl)-L-cysteine (DCVC), potently inhibits innate immune responses to GBS in human FM in culture. Despite these provocative findings, little is known about how DCVC and other toxicants modify the risk for pathogenic infection of FM. Infection of the gestational tissues (FM and placenta) is a leading known cause of PTB, therefore toxicant compromise of FM ability to fight off infectious microorganisms could significantly contribute to PTB risk. This Perspective provides the current status of understanding of toxicant-pathogen interactions in FM, highlighting knowledge gaps, challenges, and opportunities for research that can advance protections for maternal and fetal health.

10.
J Clin Invest ; 128(5): 1985-1999, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29629904

RESUMEN

Thirteen percent of pregnancies result in preterm birth or stillbirth, accounting for fifteen million preterm births and three and a half million deaths annually. A significant cause of these adverse pregnancy outcomes is in utero infection by vaginal microorganisms. To establish an in utero infection, vaginal microbes enter the uterus by ascending infection; however, the mechanisms by which this occurs are unknown. Using both in vitro and murine models of vaginal colonization and ascending infection, we demonstrate how a vaginal microbe, group B streptococcus (GBS), which is frequently associated with adverse pregnancy outcomes, uses vaginal exfoliation for ascending infection. GBS induces vaginal epithelial exfoliation by activation of integrin and ß-catenin signaling. However, exfoliation did not diminish GBS vaginal colonization as reported for other vaginal microbes. Rather, vaginal exfoliation increased bacterial dissemination and ascending GBS infection, and abrogation of exfoliation reduced ascending infection and improved pregnancy outcomes. Thus, for some vaginal bacteria, exfoliation promotes ascending infection rather than preventing colonization. Our study provides insight into mechanisms of ascending infection by vaginal microbes.


Asunto(s)
Células Epiteliales/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Vagina/inmunología , Vaginosis Bacteriana/inmunología , Animales , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Ratones , Ratones Noqueados , Infecciones Estreptocócicas/patología , Vagina/microbiología , Vagina/patología , Vaginosis Bacteriana/microbiología , Vaginosis Bacteriana/patología
11.
Nat Med ; 24(3): 368-374, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29400709

RESUMEN

Zika virus (ZIKV) is a flavivirus with teratogenic effects on fetal brain, but the spectrum of ZIKV-induced brain injury is unknown, particularly when ultrasound imaging is normal. In a pregnant pigtail macaque (Macaca nemestrina) model of ZIKV infection, we demonstrate that ZIKV-induced injury to fetal brain is substantial, even in the absence of microcephaly, and may be challenging to detect in a clinical setting. A common and subtle injury pattern was identified, including (i) periventricular T2-hyperintense foci and loss of fetal noncortical brain volume, (ii) injury to the ependymal epithelium with underlying gliosis and (iii) loss of late fetal neuronal progenitor cells in the subventricular zone (temporal cortex) and subgranular zone (dentate gyrus, hippocampus) with dysmorphic granule neuron patterning. Attenuation of fetal neurogenic output demonstrates potentially considerable teratogenic effects of congenital ZIKV infection even without microcephaly. Our findings suggest that all children exposed to ZIKV in utero should receive long-term monitoring for neurocognitive deficits, regardless of head size at birth.


Asunto(s)
Feto/virología , Complicaciones Infecciosas del Embarazo/fisiopatología , Infección por el Virus Zika/virología , Virus Zika/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Feto/fisiopatología , Humanos , Macaca nemestrina/virología , Microcefalia/diagnóstico por imagen , Microcefalia/fisiopatología , Microcefalia/virología , Neurogénesis/genética , Embarazo , Complicaciones Infecciosas del Embarazo/diagnóstico por imagen , Complicaciones Infecciosas del Embarazo/virología , Virus Zika/genética , Infección por el Virus Zika/genética , Infección por el Virus Zika/fisiopatología
12.
mBio ; 7(3)2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27353757

RESUMEN

UNLABELLED: Preterm birth increases the risk of adverse birth outcomes and is the leading cause of neonatal mortality. A significant cause of preterm birth is in utero infection with vaginal microorganisms. These vaginal microorganisms are often recovered from the amniotic fluid of preterm birth cases. A vaginal microorganism frequently associated with preterm birth is group B streptococcus (GBS), or Streptococcus agalactiae However, the molecular mechanisms underlying GBS ascension are poorly understood. Here, we describe the role of the GBS hyaluronidase in ascending infection and preterm birth. We show that clinical GBS strains associated with preterm labor or neonatal infections have increased hyaluronidase activity compared to commensal strains obtained from rectovaginal swabs of healthy women. Using a murine model of ascending infection, we show that hyaluronidase activity was associated with increased ascending GBS infection, preterm birth, and fetal demise. Interestingly, hyaluronidase activity reduced uterine inflammation but did not impact placental or fetal inflammation. Our study shows that hyaluronidase activity enables GBS to subvert uterine immune responses, leading to increased rates of ascending infection and preterm birth. These findings have important implications for the development of therapies to prevent in utero infection and preterm birth. IMPORTANCE: GBS are a family of bacteria that frequently colonize the vagina of pregnant women. In some cases, GBS ascend from the vagina into the uterine space, leading to fetal injury and preterm birth. Unfortunately, little is known about the mechanisms underlying ascending GBS infection. In this study, we show that a GBS virulence factor, HylB, shows higher activity in strains isolated from cases of preterm birth than those isolates from rectovaginal swabs of healthy women. We discovered that GBS rely on HylB to avoid immune detection in uterine tissue, but not placental tissue, which leads to increased rates of fetal injury and preterm birth. These studies provide novel insight into the underlying mechanisms of ascending infection.


Asunto(s)
Hialuronoglucosaminidasa/metabolismo , Complicaciones Infecciosas del Embarazo/microbiología , Nacimiento Prematuro/microbiología , Infecciones del Sistema Genital/microbiología , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/enzimología , Líquido Amniótico/microbiología , Femenino , Humanos , Recién Nacido , Inflamación , Placenta/inmunología , Placenta/microbiología , Embarazo , Infecciones del Sistema Genital/complicaciones , Infecciones Estreptocócicas/sangre , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/inmunología , Streptococcus agalactiae/aislamiento & purificación , Útero/inmunología , Útero/microbiología , Vagina/microbiología , Factores de Virulencia
13.
Sci Immunol ; 1(4)2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27819066

RESUMEN

Preterm birth is a leading cause of neonatal morbidity and mortality. Although microbial invasion of the amniotic cavity (MIAC) is associated with the majority of early preterm births, the temporal events that occur during MIAC and preterm labor are not known. Group B Streptococci (GBS) are ß-hemolytic, gram-positive bacteria, which commonly colonize the vagina but have been recovered from the amniotic fluid in preterm birth cases. To understand temporal events that occur during MIAC, we utilized a unique chronically catheterized nonhuman primate model that closely emulates human pregnancy. This model allows monitoring of uterine contractions, timing of MIAC and immune responses during pregnancy-associated infections. Here, we show that adverse outcomes such as preterm labor, MIAC, and fetal sepsis were observed more frequently during infection with hemolytic GBS when compared to nonhemolytic GBS. Although MIAC was associated with systematic progression in chorioamnionitis beginning with chorionic vasculitis and progressing to neutrophilic infiltration, the ability of the GBS hemolytic pigment toxin to induce neutrophil cell death and subvert killing by neutrophil extracellular traps (NETs) in placental membranes in vivo facilitated MIAC and fetal injury. Furthermore, compared to maternal neutrophils, fetal neutrophils exhibit decreased neutrophil elastase activity and impaired phagocytic functions to GBS. Collectively, our studies demonstrate how a unique bacterial hemolytic lipid toxin enables GBS to circumvent neutrophils and NETs in placental membranes to induce fetal injury and preterm labor.

14.
Nat Med ; 22(11): 1256-1259, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27618651

RESUMEN

We describe the development of fetal brain lesions after Zika virus (ZIKV) inoculation in a pregnant pigtail macaque. Periventricular lesions developed within 10 d and evolved asymmetrically in the occipital-parietal lobes. Fetal autopsy revealed ZIKV in the brain and significant cerebral white matter hypoplasia, periventricular white matter gliosis, and axonal and ependymal injury. Our observation of ZIKV-associated fetal brain lesions in a nonhuman primate provides a model for therapeutic evaluation.


Asunto(s)
Encéfalo/diagnóstico por imagen , Feto/diagnóstico por imagen , Complicaciones Infecciosas del Embarazo/diagnóstico por imagen , Infección por el Virus Zika/diagnóstico por imagen , Animales , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/virología , Colina/metabolismo , Creatina/metabolismo , Ecoencefalografía , Femenino , Feto/metabolismo , Feto/patología , Feto/virología , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Inositol/metabolismo , Macaca nemestrina , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Embarazo , Complicaciones Infecciosas del Embarazo/metabolismo , Complicaciones Infecciosas del Embarazo/patología , ARN Viral/metabolismo , Ultrasonografía Prenatal , Virus Zika/genética , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/patología
15.
Am J Reprod Immunol ; 73(3): 263-72, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25263616

RESUMEN

PROBLEM: Group B Streptococcus (GBS) is a leading cause of neonatal morbidity and mortality. We tested the hypothesis that the choriodecidua plays a role in GBS-stimulated human beta defensin(HBD)-2 increases in amnion cells through a secreted factor of choriodecidual origin. METHOD OF STUDY: Human amnion epithelial cells were treated with choriodecidual GBS-conditioned medium, live GBS, lipoteichoic acid (LTA), or lipopolysaccharide (LPS), with and without IL-1 inhibitors. RESULTS: Choriodecidual tissue punches released IL-1α and IL-1ß in response to GBS and this medium significantly stimulated release of HBD-2 by amnion cell cultures. Inhibitors of IL-1 significantly impaired the release of HBD-2 from amnion cells treated with GBS choriodecidual conditioned medium. Direct stimulation of amnion cells with GBS, LTA, or LPS did not increase HBD-2 release. CONCLUSION: Paracrine signaling involving IL-1 of choriodecidual origin is likely a critical driver for amnion HBD-2 increases in response to GBS infection of extraplacental membranes.


Asunto(s)
Amnios/metabolismo , Corion/metabolismo , Decidua/metabolismo , Interleucina-1alfa/fisiología , Interleucina-1beta/fisiología , Streptococcus agalactiae/fisiología , beta-Defensinas/biosíntesis , Amnios/efectos de los fármacos , Células Cultivadas , Corion/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Decidua/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Inmunoglobulina A/farmacología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1alfa/antagonistas & inhibidores , Interleucina-1alfa/metabolismo , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Comunicación Paracrina , Embarazo , Ácidos Teicoicos/farmacología , beta-Defensinas/genética , beta-Defensinas/metabolismo
16.
Reprod Toxicol ; 52: 1-6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25653212

RESUMEN

Extraplacental membranes define the gestational compartment and provide a barrier to infectious microorganisms ascending the gravid female reproductive tract. We tested the hypothesis that bioactive metabolites of trichloroethylene (TCE) decrease pathogen-stimulated innate immune response of extraplacental membranes. Extraplacental membranes were cultured for 4, 8, and 24h with the TCE metabolites trichloroacetate (TCA) or S-(1,2-dichlorovinyl)-l-cysteine (DCVC) in the absence or presence of lipoteichoic acid (LTA) or lipopolysaccharide (LPS) to simulate infection. In addition, membranes were cocultured with DCVC and Group B Streptococcus (GBS). DCVC (5-50µM) significantly inhibited LTA-, LPS-, and GBS-stimulated cytokine release from tissue cultures as early as 4h (P≤0.05). In contrast, TCA (up to 500µM) did not inhibit LTA-stimulated cytokine release from tissue punches. Because cytokines are important mediators for host response to infectious microorganisms these findings suggest that TCE exposure could potentially modify susceptibility to infection during pregnancy.


Asunto(s)
Cisteína/análogos & derivados , Membranas Extraembrionarias/inmunología , Inmunidad/efectos de los fármacos , Streptococcus agalactiae/inmunología , Ácido Tricloroacético/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Corion/inmunología , Cisteína/farmacología , Decidua/inmunología , Resistencia a la Enfermedad/efectos de los fármacos , Femenino , Humanos , Lipopolisacáridos/farmacología , Embarazo , Infecciones Estreptocócicas/inmunología , Ácidos Teicoicos/farmacología , Técnicas de Cultivo de Tejidos , Tricloroetileno/metabolismo
17.
Sci Adv ; 1(6): e1400225, 2015 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-26425734

RESUMEN

Ascending infection of microbes from the lower genital tract into the amniotic cavity increases the risk of preterm birth, stillbirth, and newborn infections. Host defenses that are critical for preventing ascending microbial infection are not completely understood. Group B Streptococcus (GBS) are Gram-positive bacteria that frequently colonize the lower genital tract of healthy women but cause severe infections during pregnancy, leading to preterm birth, stillbirth, or early-onset newborn infections. We recently described that the GBS pigment is hemolytic, and increased pigment expression promotes GBS penetration of human placenta. Here, we show that the GBS hemolytic pigment/lipid toxin and hyperpigmented GBS strains induce mast cell degranulation, leading to the release of preformed and proinflammatory mediators. Mast cell-deficient mice exhibit enhanced bacterial burden, decreased neutrophil mobilization, and decreased immune responses during systemic GBS infection. In a vaginal colonization model, hyperpigmented GBS strains showed increased persistence in mast cell-deficient mice compared to mast cell-proficient mice. Consistent with these observations, fewer rectovaginal GBS isolates from women in their third trimester of pregnancy were hyperpigmented/hyperhemolytic. Our work represents the first example of a bacterial hemolytic lipid that induces mast cell degranulation and emphasizes the role of mast cells in limiting genital colonization by hyperpigmented GBS.

18.
EMBO Mol Med ; 7(4): 488-505, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25750210

RESUMEN

Group B streptococci (GBS) are Gram-positive bacteria that cause infections in utero and in newborns. We recently showed that the GBS pigment is hemolytic and increased pigment production promotes bacterial penetration of human placenta. However, mechanisms utilized by the hemolytic pigment to induce host cell lysis and the consequence on fetal injury are not known. Here, we show that the GBS pigment induces membrane permeability in artificial lipid bilayers and host cells. Membrane defects induced by the GBS pigment trigger K(+) efflux leading to osmotic lysis of red blood cells or pyroptosis in human macrophages. Macrophages lacking the NLRP3 inflammasome recovered from pigment-induced cell damage. In a murine model of in utero infection, hyperpigmented GBS strains induced fetal injury in both an NLRP3 inflammasome-dependent and NLRP3 inflammasome-independent manner. These results demonstrate that the dual mechanism of action of the bacterial pigment/lipid toxin leading to hemolysis or pyroptosis exacerbates fetal injury and suggest that preventing both activities of the hemolytic lipid is likely critical to reduce GBS fetal injury and preterm birth.


Asunto(s)
Toxinas Bacterianas , Permeabilidad de la Membrana Celular , Enfermedades Fetales , Lípidos de la Membrana , Piroptosis/inmunología , Infecciones Estreptocócicas , Streptococcus agalactiae , Animales , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/toxicidad , Línea Celular Tumoral , Femenino , Enfermedades Fetales/inmunología , Enfermedades Fetales/microbiología , Enfermedades Fetales/patología , Humanos , Masculino , Lípidos de la Membrana/inmunología , Lípidos de la Membrana/toxicidad , Ratones , Ratones Noqueados , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/patología , Streptococcus agalactiae/inmunología , Streptococcus agalactiae/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA