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










Base de datos
Intervalo de año de publicación
2.
Front Virol ; 22022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35573818

RESUMEN

Zika virus (ZIKV) infection during pregnancy causes serious adverse outcomes to the developing fetus, including fetal loss and birth defects known as congenital Zika syndrome (CZS). The mechanism by which ZIKV infection causes these adverse outcomes and specifically, the interplay between the maternal immune response and ZIKV replication has yet to be fully elucidated. Using an immunocompetent mouse model of transplacental ZIKV transmission and adverse pregnancy outcomes, we have previously shown that Asian lineage ZIKV disrupts placental morphology and induces elevated secretion of IL-1ß. In the current manuscript, we characterized placental damage and inflammation during in utero African lineage ZIKV infection. Within 48 hours after ZIKV infection at embryonic day 10, viral RNA was detected in placentas and fetuses from ZIKA infected dams, which corresponded with placental damage and reduced fetal viability as compared with mock infected dams. Dams infected with ZIKV had reduced proportions of trophoblasts and endothelial cells and disrupted placental morphology compared to mock infected dams. While placental IL-1ß was increased in the placenta, but not the spleen, within 3 hours post infection, this was not caused by activation of the NLRP3 inflammasome. Using bulk mRNAseq from placentas of ZIKV and mock infected dams, ZIKV infection caused profound downregulation of the transcriptional activity of genes that may underly tissue morphology, neurological development, metabolism, cell signaling and inflammation, illustrating that in utero ZIKV infections causes disruption of pathways associated with CZS in our model.

3.
Front Bioeng Biotechnol ; 10: 819593, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35155393

RESUMEN

Intrauterine inflammation (IUI) is the primary cause of spontaneous preterm birth and predisposes neonates to long-term sequelae, including adverse neurological outcomes. N-acetyl-L-cysteine (NAC) is the amino acid L-cysteine derivative and a precursor to the antioxidant glutathione (GSH). NAC is commonly used clinically as an antioxidant with anti-inflammatory properties. Poor bioavailability and high protein binding of NAC necessitates the use of high doses resulting in side effects including nausea, vomiting, and gastric disruptions. Therefore, dendrimer-based therapy can specifically target the drug to the cells involved in inflammation, reducing side effects with efficacy at much lower doses than the free drug. Towards development of the new therapies for the treatment of maternal inflammation, we successfully administered dendrimer-based N-Acetyl Cysteine (DNAC) in an animal model of IUI to reduce preterm birth and perinatal inflammatory response. This study explored the associated immune mechanisms of DNAC treatment on placental macrophages following IUI, especially on M1/M2 type macrophage polarization. Our results demonstrated that intraperitoneal maternal DNAC administration significantly reduced the pro-inflammatory cytokine mRNA of Il1ß and Nos2, and decreased CD45+ leukocyte infiltration in the placenta following IUI. Furthermore, we found that DNAC altered placental immune profile by stimulating macrophages to change to the M2 phenotype while decreasing the M1 phenotype, thus suppressing the inflammatory responses in the placenta. Our study provides evidence for DNAC therapy to alleviate IUI via the maintenance of macrophage M1/M2 imbalance in the placenta.

4.
J Reprod Immunol ; 147: 103360, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34390899

RESUMEN

Preterm birth (PTB) is considered to be one of the most frequent causes of neonatal death. Prompt and effective measures to predict adverse fetal outcome following PTB are urgently needed. Placenta macrophages are a critical immune cell population during pregnancy, phenotypically divided into M1 and M2 subsets. An established mouse model of intrauterine inflammation (IUI) was applied. Placenta (labyrinth) and corresponding fetal brain were harvested within 24 hours post injection (hpi). Flow cytometry, Western blot, real-time qPCR, and regular histology were utilized to examine the cytokines, macrophage polarization, and sex-specificity. Placental exposure to LPS led to significantly reduced labyrinth thickness compared to PBS-exposed controls as early as 3 hpi, accompanied by apoptosis and necrosis. Pro-inflammatory M1 markers, Il-1ß, and iNOS, and anti-inflammatory M2 marker Il-10 increased significantly in placentas exposed to IUI. Analysis of flow cytometry revealed that fetal macrophages (Hofbauer cell, HBCs) were mostly M1-like and that maternal inter-labyrinth macrophages (MIM) were M2-like in their features in IUI. Male fetuses displayed significantly decreased M2-like features in HBCs at 3 and 6 hpi, while female fetuses showed significant increase in M2-like features in MIM at 3 and 6 hpi. Furthermore, there was a significant correlation between the frequency of HBCs and corresponding microglial marker expression at 3 and 6 hpi. Placental macrophages demonstrated sex-specific features in response to IUI. Specifically, HBCs may be a potential biomarker for fetal brain injury at preterm birth.


Asunto(s)
Macrófagos/inmunología , Microglía/inmunología , Enfermedades Neuroinflamatorias/diagnóstico , Placenta/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Microglía/patología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/patología , Placenta/citología , Placenta/patología , Embarazo , Pronóstico , Factores Sexuales
5.
PLoS Negl Trop Dis ; 14(10): e0008707, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33091001

RESUMEN

In utero Zika virus (ZIKV; family Flaviviridae) infection causes a distinct pattern of birth defects and disabilities in the developing fetus and neonate that has been termed congenital zika syndrome (CZS). Over 8,000 children were affected by the 2016 to 2017 ZIKV outbreak in the Americas, many of whom developed CZS as a result of in utero exposure. To date, there is no consensus about how ZIKV causes CZS; animal models, however, are providing mechanistic insights. Using nonhuman primates, immunocompromised mice, immunocompetent mice, and other animal models (e.g., pigs, sheep, guinea pigs, and hamsters), studies are showing that maternal immunological responses, placental infection and inflammation, as well as viral genetic factors play significant roles in predicting the downstream consequences of in utero ZIKV infection on the development of CZS in offspring. There are thousands of children suffering from adverse consequences of CZS. Therefore, the animal models developed to study ZIKV-induced adverse outcomes in offspring could provide mechanistic insights into how other viruses, including influenza and hepatitis C viruses, impact placental viability and fetal growth to cause long-term adverse outcomes in an effort to identify therapeutic treatments.


Asunto(s)
Modelos Animales de Enfermedad , Infección por el Virus Zika/congénito , Virus Zika/fisiología , Animales , Femenino , Embarazo , Infección por el Virus Zika/virología
6.
J Virol ; 95(2)2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33115865

RESUMEN

Viral infections during pregnancy lead to a spectrum of maternal and fetal outcomes, ranging from asymptomatic disease to more critical conditions presenting with severe maternal morbidity, stillbirth, preterm birth, intrauterine growth restriction, and fetal congenital anomalies, either apparent at birth or later in life. In this article, we review the pathogenesis of several viral infections that are particularly relevant in the context of pregnancy and intrauterine inflammation. Understanding the diverse mechanisms employed by viral pathogens as well as the repertoire of immune responses induced in the mother may help to establish novel therapeutic options to attenuate changes in the maternal-fetal interface and prevent adverse pregnancy outcomes.


Asunto(s)
Inflamación/inmunología , Complicaciones Infecciosas del Embarazo/virología , Virosis/virología , Virus/patogenicidad , Femenino , Humanos , Recién Nacido , Placenta/inmunología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/patología , Resultado del Embarazo , Virosis/inmunología , Virosis/patología , Virus/clasificación , Virus/inmunología
7.
J Pineal Res ; 69(3): e12687, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32737901

RESUMEN

Inflammation is associated with injury to immature lungs, and melatonin administration to preterm newborns with acute respiratory distress improves pulmonary outcomes. We hypothesized that maternally administered melatonin may reduce inflammation, oxidative stress, and structural injury in fetal lung and help fetal lung maturation in a mouse model of intrauterine inflammation (IUI). Mice were randomized to the following groups: control (C), melatonin (M), lipopolysaccharide (LPS; a model of IUI) (L), and LPS with melatonin (ML). Pro-inflammatory cytokines, components of the Hippo pathway, and Yap1/Taz were analyzed in the fetal lung at E18 by real-time RT-qPCR. Confirmatory histochemistry and immunohistochemical analyses (surfactant protein B, vimentin, HIF-1ß, and CXCR2) were performed. The gene expression of IL1ß in the fetal lung was significantly increased in L compared to C, M, and ML. Taz expression was significantly decreased in L compared to C and M. Taz gene expression in L was significantly decreased compared with those in ML. Immunohistochemical analyses showed that the expression of HIF-1ß and CXCR2 was significantly increased in L compared to C, M, and ML. The area of surfactant protein B and vimentin were significantly decreased in L than C, M, or ML in the fetal and neonatal lung. Antenatal maternally administered melatonin appears to prevent fetal lung injury induced by IUI and to help lung maturation. The results from this study results suggest that melatonin could serve as a novel safe preventive and/or therapeutic medicine for preventing fetal lung injury from IUI and for improving lung maturation in prematurity.


Asunto(s)
Enfermedades Fetales , Feto/embriología , Lesión Pulmonar , Pulmón/embriología , Melatonina/farmacología , Animales , Femenino , Enfermedades Fetales/inducido químicamente , Enfermedades Fetales/prevención & control , Inflamación/inducido químicamente , Inflamación/embriología , Inflamación/prevención & control , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/embriología , Lesión Pulmonar/prevención & control , Ratones , Embarazo
8.
Am J Reprod Immunol ; 84(1): e13248, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32306461

RESUMEN

PROBLEM: Systemic maternal inflammation is associated with adverse neonatal sequelae. We tested the hypothesis that IL-1ß is a key inflammatory regulator of adverse pregnancy outcomes. METHOD OF STUDY: Pregnant mice were treated with intraperitoneal injections of IL-1ß (0, 0.1, 0.5, or 1 µg) from embryonic day (E)14 to E17. Placenta and fetal brains were harvested and analyzed for morphologic changes and IL-1ß signaling markers. RESULTS: As compared with non-treated dams, maternal injections with IL-1ß resulted in increased p-NF-κB and caspase-1 in placentas and fetal brains, but not consistently in spleens, suggesting induction of intrinsic IL-1ß production. These findings were confirmed by increased levels of IL-1ß in the placentas of the IL-1ß-treated dams. Systemic treatment of dams with IL-1ß suppressed Stat1 signaling. Maternal inflammation caused by IL-1ß treatment reduced fetal viability to 80.6% and 58.9%, in dams treated with either 0.5 or 1 µg of IL-1ß, respectively. In the placentas, there was an IL-1ß dose-dependent distortion of the labyrinth structure, decreased numbers of mononuclear trophoblast giant cells, and reduced proportions of endothelial cells as compared to placentas from control dams. In fetal brains collected at E17, there was an IL-1ß dose-dependent reduction in cortical neuronal morphology. CONCLUSION: This work demonstrates that systemic IL-1ß injection causes dose-dependent structural and functional changes in the placenta and fetal brain.


Asunto(s)
Encéfalo/inmunología , Inflamación/inmunología , Placenta/inmunología , Complicaciones del Embarazo/inmunología , Animales , Células Cultivadas , Femenino , Feto , Regulación de la Expresión Génica , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/inmunología , Ratones , FN-kappa B/metabolismo , Embarazo , Transducción de Señal
9.
Dev Neurobiol ; 80(5-6): 149-159, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32333505

RESUMEN

Fetuses exposed to an inflammatory environment are predisposed to long-term adverse neurological outcomes. However, the mechanism by which intrauterine inflammation (IUI) is responsible for abnormal fetal brain development is not fully understood. The mechanistic target of rapamycin (mTOR) signaling pathway is closely associated with fetal brain development. We hypothesized that mTOR signaling might be involved in fetal brain injury and malformation when fetuses are exposed to the IUI environment. A well-established IUI model was utilized by intrauterine injection of lipopolysaccharide (LPS) to explore the effect of IUI on mTOR signaling in mouse fetal brains. We found that microglia activation in LPS fetal brains was increased, as demonstrated by elevated Iba-1 protein level and immunofluorescence density. LPS fetal brains also showed reduced neuronal cell counts, decreased cell proliferation demonstrated by low Ki67-positive density, and elevated neuron apoptosis evidenced by high expression of cleaved Caspase 3. Furthermore, we found that mTOR signaling in LPS fetal brains was elevated at 2 hr after LPS treatment, declined at 6 hr and showed overall inhibition at 24 hr. In summary, our study revealed that LPS-induced IUI leads to increased activation of microglia cells, neuronal damage, and dynamic alterations in mTOR signaling in the mouse fetal brain. Our findings indicate that abnormal changes in mTOR signaling may underlie the development of future neurological complications in offspring exposed to prenatal IUI.


Asunto(s)
Encefalopatías , Corteza Cerebral , Desarrollo Fetal/fisiología , Enfermedades Fetales , Inflamación , Microglía , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Encefalopatías/inmunología , Encefalopatías/metabolismo , Encefalopatías/patología , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal/inmunología , Enfermedades Fetales/inmunología , Enfermedades Fetales/metabolismo , Enfermedades Fetales/patología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Ratones , Microglía/inmunología , Microglía/metabolismo , Embarazo , Transducción de Señal/inmunología
10.
Biol Reprod ; 101(5): 1046-1055, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31403169

RESUMEN

Maternal inflammation (MI) is associated with many adverse perinatal outcomes. The placenta plays a vital role in mediating maternal-fetal resource allocation. Studies have shown that MI contributes to placental dysfunction, which then leads to adverse birth outcomes and high health risks throughout childhood. Placental mammalian target of rapamycin complex 1 (mTORC1) signaling pathway links maternal nutrient availability to fetal growth; however, the impact of MI on mTORC1 signaling in the placenta remains unclear. In this study, we sought to explore the changes of mTORC1 signaling in the mouse placenta at late gestation by using two models of MI employing lipopolysaccharide (LPS) and interleukin-1ß (IL-1ß) to mimic acute (aMI) and sub-chronic (cMI) inflammatory states, respectively. We determined placental mTORC1 activity by measuring the activity of mTORC1 downstream molecules, including S6k, 4Ebp1, and rpS6. In the aMI model, we found that mTORC1 activity was significantly decreased in the placental decidual and junctional zone at 2 and 6 h after LPS surgery, respectively; however, mTORC1 activity was significantly increased in the placental labyrinth zone at 2, 6, and 24 h after LPS treatment, respectively. In the cMI model, we observed that mTORC1 activity was increased only in the placental labyrinth zone after consecutive IL-1ß exposure. Our study reveals that different parts of the mouse placenta react differently to MI, leading to variable mTORC1 activity throughout the placenta. This suggests that different downstream molecules of mTORC1 from different parts of the mouse placenta may be used in clinical research to monitor the fetal well-being during MI.


Asunto(s)
Inflamación/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Placenta/metabolismo , Animales , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Interleucina-1beta/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Fosforilación , Embarazo , Distribución Aleatoria , Proteína S6 Ribosómica/genética , Proteína S6 Ribosómica/metabolismo
11.
J Reprod Immunol ; 133: 52-62, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31280130

RESUMEN

PROBLEM: Exposure to systemic maternal inflammation (i.e., maternal sepsis, influenza, human immunodeficiency virus, or pyelonephritis) and intrauterine (IU) inflammation (i.e., chorioamnionitis or preterm labor) have been associated with adverse perinatal sequelae. Whether systemic and localized inflammation leading to adverse outcomes have similar placental mechanisms remain unclear. METHOD OF STUDY: We conducted a study by murine modeling systemic and localized IU inflammation with injections of either intraperitoneal (IP) or IU interleukin-1ß (IL-1ß) and compared fetoplacental hemodynamic changes, cytokine/chemokine expression, and fetal loss. RESULTS: IU IL-1ß exposure reduced offspring survival by 31.1% and IP IL-1ß exposure by 34.5% when compared with control pups. Despite this similar outcome in offspring survival, Doppler analysis revealed a stark difference: IU group displayed worsened fetoplacental hemodynamic changes while no differences were found between IP and control groups. While both IU and IP groups had increases in pro-inflammatory cytokines and chemokines, specific gene expression trends differed between the two groups, once again highlighting their mechanistic differences. CONCLUSION: While both IP and IU IL-1ß exposure similarly affected pup survival, only IU inflammation resulted in fetoplacental hemodynamic changes. We speculate that exposure to maternal systemic and IU inflammation plays a key role in fetal injury by utilizing different placental inflammatory pathways and mechanisms.


Asunto(s)
Corioamnionitis/inmunología , Intercambio Materno-Fetal/inmunología , Placenta/inmunología , Nacimiento Prematuro/inmunología , Animales , Corioamnionitis/diagnóstico por imagen , Corioamnionitis/mortalidad , Corioamnionitis/patología , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Feto/diagnóstico por imagen , Feto/inmunología , Humanos , Interleucina-1beta/administración & dosificación , Interleucina-1beta/inmunología , Ratones , Placenta/patología , Circulación Placentaria/inmunología , Embarazo , Nacimiento Prematuro/mortalidad , Nacimiento Prematuro/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Tasa de Supervivencia , Ultrasonografía Doppler
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA