Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
mBio ; 12(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33402537

RESUMEN

Invasive bacterial infections during pregnancy are a major risk factor for preterm birth, stillbirth, and fetal injury. Group B streptococci (GBS) are Gram-positive bacteria that asymptomatically colonize the lower genital tract but infect the amniotic fluid and induce preterm birth or stillbirth. Experimental models that closely emulate human pregnancy are pivotal for the development of successful strategies to prevent these adverse pregnancy outcomes. Using a unique nonhuman primate model that mimics human pregnancy and informs temporal events surrounding amniotic cavity invasion and preterm labor, we show that the animals inoculated with hyaluronidase (HylB)-expressing GBS consistently exhibited microbial invasion into the amniotic cavity, fetal bacteremia, and preterm labor. Although delayed cytokine responses were observed at the maternal-fetal interface, increased prostaglandin and matrix metalloproteinase levels in these animals likely mediated preterm labor. HylB-proficient GBS dampened reactive oxygen species production and exhibited increased resistance to neutrophils compared to an isogenic mutant. Together, these findings demonstrate how a bacterial enzyme promotes GBS amniotic cavity invasion and preterm labor in a model that closely resembles human pregnancy.IMPORTANCE Group B streptococci (GBS) are bacteria that commonly reside in the female lower genital tract as asymptomatic members of the microbiota. However, during pregnancy, GBS can infect tissues at the maternal-fetal interface, leading to preterm birth, stillbirth, or fetal injury. Understanding how GBS evade host defenses during pregnancy is key to developing improved preventive therapies for these adverse outcomes. In this study, we used a unique nonhuman primate model to show that an enzyme secreted by GBS, hyaluronidase (HylB) promotes bacterial invasion into the amniotic cavity and fetus. Although delayed immune responses were seen at the maternal-fetal interface, animals infected with hyaluronidase-expressing GBS exhibited premature cervical ripening and preterm labor. These observations reveal that HylB is a crucial GBS virulence factor that promotes bacterial invasion and preterm labor in a pregnancy model that closely emulates human pregnancy. Therefore, hyaluronidase inhibitors may be useful in therapeutic strategies against ascending GBS infection.


Asunto(s)
Hialuronoglucosaminidasa/metabolismo , Neutrófilos/inmunología , Trabajo de Parto Prematuro/inmunología , Infecciones Estreptocócicas/inmunología , Streptococcus agalactiae/metabolismo , Líquido Amniótico/microbiología , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Hialuronoglucosaminidasa/genética , Inflamación , Pulmón/microbiología , Pulmón/patología , Macaca nemestrina , Neutrófilos/microbiología , Embarazo , Nacimiento Prematuro , Primates , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/enzimología , Streptococcus agalactiae/genética , Streptococcus agalactiae/inmunología
2.
Front Immunol ; 11: 770, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425945

RESUMEN

Leukocyte activation within the chorioamniotic membranes is strongly associated with inflammation and preterm labor (PTL). We hypothesized that prophylaxis with a broad-spectrum chemokine inhibitor (BSCI) would downregulate the inflammatory microenvironment induced by Group B Streptococcus (GBS, Streptococcus agalactiae) to suppress PTL and microbial invasion of the amniotic cavity (MIAC). To correlate BSCI administration with PTL and MIAC, we used a unique chronically catheterized non-human primate model of Group B Streptococcus (GBS)-induced PTL. In the early third trimester (128-138 days gestation; ~29-32 weeks human pregnancy), animals received choriodecidual inoculations of either: (1) saline (N = 6), (2) GBS, 1-5 × 108 colony forming units (CFU)/ml; N = 5), or (3) pre-treatment and daily infusions of a BSCI (10 mg/kg intravenous and intra-amniotic) with GBS (1-5 × 108 CFU/ml; N = 4). We measured amniotic cavity pressure (uterine contraction strength) and sampled amniotic fluid (AF) and maternal blood serially and cord blood at delivery. Cesarean section was performed 3 days post-inoculation or earlier for PTL. Data analysis used Fisher's exact test, Wilcoxon rank sum and one-way ANOVA with Bonferroni correction. Saline inoculation did not induce PTL or infectious sequelae. In contrast, GBS inoculation typically induced PTL (4/5, 80%), MIAC and fetal bacteremia (3/5; 60%). Remarkably, PTL did not occur in the BSCI+GBS group (0/4, 0%; p = 0.02 vs. GBS), despite MIAC and fetal bacteremia in all cases (4/4; 100%). Compared to the GBS group, BSCI prophylaxis was associated with significantly lower cytokine levels including lower IL-8 in amniotic fluid (p = 0.03), TNF-α in fetal plasma (p < 0.05), IFN-α and IL-7 in the fetal lung (p = 0.02) and IL-18, IL-2, and IL-7 in the fetal brain (p = 0.03). Neutrophilic chorioamnionitis was common in the BSCI and GBS groups, but was more severe in the BSCI+GBS group with greater myeloperoxidase staining (granulocyte marker) in the amnion and chorion (p < 0.05 vs. GBS). Collectively, these observations indicate that blocking the chemokine response to infection powerfully suppressed uterine contractility, PTL and the cytokine response, but did not prevent MIAC and fetal pneumonia. Development of PTL immunotherapies should occur in tandem with evaluation for AF microbes and consideration for antibiotic therapy.


Asunto(s)
Líquido Amniótico/microbiología , Quimiocinas/antagonistas & inhibidores , Trabajo de Parto Prematuro/prevención & control , Streptococcus agalactiae/patogenicidad , Animales , Animales Recién Nacidos , Cesárea , Citocinas/análisis , Femenino , Macrófagos/fisiología , Morbilidad , Neutrófilos/efectos de los fármacos , Neutrófilos/fisiología , Embarazo , Primates , Infecciones Estreptocócicas/complicaciones
3.
Nat Commun ; 11(1): 1502, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32198389

RESUMEN

Although certain microbial lipids are toxins, the structural features important for cytotoxicity remain unknown. Increased functional understanding is essential for developing therapeutics against toxic microbial lipids. Group B Streptococci (GBS) are bacteria associated with preterm births, stillbirths, and severe infections in neonates and adults. GBS produce a pigmented, cytotoxic lipid, known as granadaene. Despite its importance to all manifestations of GBS disease, studies towards understanding granadaene's toxic activity are hindered by its instability and insolubility in purified form. Here, we report the synthesis and screening of lipid derivatives inspired by granadaene, which reveal features central to toxin function, namely the polyene chain length. Furthermore, we show that vaccination with a non-toxic synthetic analog confers the production of antibodies that inhibit granadaene-mediated hemolysis ex vivo and diminish GBS infection in vivo. This work provides unique structural and functional insight into granadaene and a strategy to mitigate GBS infection, which will be relevant to other toxic lipids encoded by human pathogens.


Asunto(s)
Hemólisis , Lípidos/química , Polienos/química , Nacimiento Prematuro/microbiología , Infecciones Estreptocócicas/metabolismo , Adulto , Animales , Linfocitos B , Toxinas Bacterianas/química , Vacunas Bacterianas , Linfocitos T CD4-Positivos , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Lípidos/inmunología , Lípidos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Polienos/inmunología , Embarazo , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/prevención & control , Streptococcus agalactiae , Vacunación
4.
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
5.
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
6.
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
7.
J Infect Dis ; 217(7): 1128-1138, 2018 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-29301010

RESUMEN

Group B streptococci (GBS) are encapsulated, ß-hemolytic bacteria that are a common cause of infections in human newborns and certain adults. Two factors important for GBS virulence are the sialic acid capsular polysaccharide that promotes immune evasion and the hemolytic pigment that induces host cell cytotoxcity. These virulence factors are often oppositely regulated by the CovR/CovS two-component system. Clinical GBS strains exhibiting hyperhemolysis and low capsule due to pathoadaptive covR/S mutations have been isolated from patients. Given the importance of capsule to GBS virulence, we predicted that a decrease or loss of capsule would attenuate the virulence of covR/S mutants. Surprisingly, hyperhemolytic GBS with low or no capsule exhibit increased virulence, intracellular persistence, and blood-brain barrier penetration, which was independent of a Trojan horse mechanism of barrier penetration. Additionally, intracellular persistence enabled both hemolytic and hyperhemolytic GBS to evade antibiotics routinely used to treat these infections. The finding that diminished capsule expression promotes GBS virulence, intracellular persistence, and antibiotic evasion has important implications for sustained antibiotic therapy and efficacy of capsule-based vaccines.


Asunto(s)
Antibacterianos/farmacología , Cápsulas Bacterianas/genética , Farmacorresistencia Bacteriana/genética , Streptococcus agalactiae/citología , Streptococcus agalactiae/patogenicidad , Animales , Barrera Hematoencefálica , Humanos , Macrófagos , Ratones , Ratones Endogámicos C57BL , Mutación , Streptococcus agalactiae/efectos de los fármacos , Streptococcus agalactiae/fisiología , Virulencia
8.
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
9.
Neurobiol Dis ; 109(Pt A): 127-136, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28986324

RESUMEN

Huntington's disease (HD) is caused by a CAG repeat expansion that encodes a polyglutamine (polyQ) expansion in the HD disease protein, huntingtin (HTT). PolyQ expansion promotes misfolding and aggregation of mutant HTT (mHTT) within neurons. The cellular pathways, including ubiquitin-dependent processes, by which mHTT is regulated remain incompletely understood. Ube2W is the only ubiquitin conjugating enzyme (E2) known to ubiquitinate substrates at their amino (N)-termini, likely favoring substrates with disordered N-termini. By virtue of its N-terminal polyQ domain, HTT has an intrinsically disordered amino terminus. In studies employing immortalized cells, primary neurons and a knock-in (KI) mouse model of HD, we tested the effect of Ube2W deficiency on mHTT levels, aggregation and neurotoxicity. In cultured cells, deficiency of Ube2W activity markedly decreases mHTT aggregate formation and increases the level of soluble monomers, while reducing mHTT-induced cytotoxicity. Consistent with this result, the absence of Ube2W in HdhQ200 KI mice significantly increases levels of soluble monomeric mHTT while reducing insoluble oligomeric species. This study sheds light on the potential function of the non-canonical ubiquitin-conjugating enzyme, Ube2W, in this polyQ neurodegenerative disease.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/enzimología , Neuronas/enzimología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Cuerpo Estriado/enzimología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Células HEK293 , Humanos , Enfermedad de Huntington/genética , Cuerpos de Inclusión/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/patología , Cultivo Primario de Células , Enzimas Ubiquitina-Conjugadoras/genética
10.
J Infect Dis ; 217(6): 983-987, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29244079

RESUMEN

Group B streptococci (GBS) are Gram-positive bacteria that are a leading cause of neonatal infections. Most invasive isolates are ß-hemolytic, and hemolytic activity is critical for GBS virulence. Although nonhemolytic GBS strains are occasionally isolated, they are often thought to be virulence attenuated. In this study, we show that a nonhemolytic GBS strain (GB37) isolated from a septic neonate exhibits hypervirulence. Substitution of tryptophan to leucine (W297L) in the sensor histidine kinase CovS results in constitutive kinase signaling, leading to decreased hemolysis and increased activity of the GBS hyaluronidase, HylB. These results describe how nonpigmented and nonhemolytic GBS strains can exhibit hypervirulence.


Asunto(s)
Infecciones Estreptocócicas/microbiología , Streptococcus agalactiae/patogenicidad , Sustitución de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Histidina Quinasa/química , Histidina Quinasa/metabolismo , Humanos , Hialuronoglucosaminidasa/metabolismo , Recién Nacido , Leucina , Ratones , Infecciones Estreptocócicas/patología , Streptococcus agalactiae/genética , Triptófano , Virulencia
11.
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.

12.
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
13.
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
14.
J Biol Chem ; 291(6): 3030-42, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26601958

RESUMEN

UBE2W ubiquitinates N termini of proteins rather than internal lysine residues, showing a preference for substrates with intrinsically disordered N termini. The in vivo functions of this intriguing E2, however, remain unknown. We generated Ube2w germ line KO mice that proved to be susceptible to early postnatal lethality without obvious developmental abnormalities. Although the basis of early death is uncertain, several organ systems manifest changes in Ube2w KO mice. Newborn Ube2w KO mice often show altered epidermal maturation with reduced expression of differentiation markers. Mirroring higher UBE2W expression levels in testis and thymus, Ube2w KO mice showed a disproportionate decrease in weight of these two organs (~50%), suggesting a functional role for UBE2W in the immune and male reproductive systems. Indeed, Ube2w KO mice displayed sustained neutrophilia accompanied by increased G-CSF signaling and testicular vacuolation associated with decreased fertility. Proteomic analysis of a vulnerable organ, presymptomatic testis, showed a preferential accumulation of disordered proteins in the absence of UBE2W, consistent with the view that UBE2W preferentially targets disordered polypeptides. These mice further allowed us to establish that UBE2W is ubiquitously expressed as a single isoform localized to the cytoplasm and that the absence of UBE2W does not alter cell viability in response to various stressors. Our results establish that UBE2W is an important, albeit not essential, protein for early postnatal survival and normal functioning of multiple organ systems.


Asunto(s)
Epidermis , Anomalías Cutáneas , Enzimas Ubiquitina-Conjugadoras , Animales , Epidermis/anomalías , Epidermis/enzimología , Epidermis/inmunología , Trastornos Leucocíticos/congénito , Trastornos Leucocíticos/enzimología , Trastornos Leucocíticos/genética , Trastornos Leucocíticos/inmunología , Masculino , Ratones , Ratones Noqueados , Anomalías Cutáneas/enzimología , Anomalías Cutáneas/genética , Anomalías Cutáneas/inmunología , Testículo/enzimología , Testículo/inmunología , Timo/enzimología , Timo/inmunología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/inmunología
15.
Neurobiol Dis ; 82: 281-288, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26141599

RESUMEN

Accumulation of mutant polyglutamine proteins in intraneuronal inclusions is a hallmark of polyglutamine diseases. Impairment of protein clearance systems and sequestration of clearance-related proteins into inclusions occur in many protein folding diseases, including polyglutamine diseases. The ubiquitin-binding and proteasome adaptor protein UBQLN2 participates in protein homeostasis and localizes to inclusions in various neurodegenerative diseases. Employing mouse models and human brain tissue of Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3), we show that UBQLN2 is selectively recruited to inclusions in HD but not SCA3. Consistent with this result, in a cell-based system mutant HTT interacts with UBQLN2 through the UBA domain while the SCA3 disease protein ATXN3, a deubiquitinating enzyme, does not interact with UBQLN2. Differential recruitment of UBQLN2 to aggregates in HD and SCA3 underscores the heterogeneity of inclusions in polyglutamine diseases and suggests that components of neuronal protein quality control may be differentially perturbed in distinct polyQ diseases.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión Intranucleares/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Ubiquitinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Ataxina-3/genética , Ataxina-3/metabolismo , Proteínas Relacionadas con la Autofagia , Encéfalo/patología , Proteínas de Ciclo Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células HEK293 , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Cuerpos de Inclusión Intranucleares/patología , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/patología , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Ubiquitinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...