Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
PLoS Pathog ; 20(2): e1011990, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38324589

RESUMO

BACKGROUND: Hofbauer cells (HBCs) and cytotrophoblasts (CTBs) are major cell populations in placenta. The indirect impact of maternal SARS-CoV-2 disease on these cells that are not directly infected has not been extensively studied. Herein, we profiled gene expression in HBCs and CTBs isolated from placentae of recovered pregnant subjects infected with SARS-CoV-2 during all trimesters of pregnancy, placentae from subjects with active infection, SARS-CoV-2 vaccinated subjects, and those who were unexposed to the virus. METHODS: Placentae were collected within 4 h post-delivery and membrane-free tissues were enzymatically digested for the isolation of HBCs and CTBs. RNA extracted from HBCs and CTBs were sequenced using 150bp paired-end reads. Differentially expressed genes (DEGs) were identified by DESeq2 package in R and enriched in GO Biological Processes, KEGG Pathway, Reactome Gene Sets, Hallmark Gene Sets, and Canonical Pathways. Protein-protein interactions among the DEGs were modelled using STRING and BioGrid. RESULTS: Pregnant subjects (n = 30) were recruited and categorized into six groups: infected with SARS-CoV-2 in i) the first (1T, n = 4), ii) second (2T, n = 5), iii) third (3T, n = 5) trimester, iv) tested positive at delivery (Delivery, n = 5), v) never infected (Control, n = 6), and vi) fully mRNA-vaccinated by delivery (Vaccinated, n = 5). Compared to the Control group, gene expression analysis showed that HBCs from infected subjects had significantly altered gene expression profiles, with the 2T group having the highest number of DEGs (1,696), followed by 3T and 1T groups (1,656 and 958 DEGs, respectively). These DEGs were enriched for pathways involved in immune regulation for host defense, including production of cytokines, chemokines, antimicrobial proteins, ribosomal assembly, neutrophil degranulation inflammation, morphogenesis, and cell migration/adhesion. Protein-protein interaction analysis mapped these DEGs with oxidative phosphorylation, translation, extracellular matrix organization, and type I interferon signaling. Only 95, 23, and 8 DEGs were identified in CTBs of 1T, 2T, and 3T groups, respectively. Similarly, 11 and 3 DEGs were identified in CTBs and HBCs of vaccinated subjects, respectively. Reassuringly, mRNA vaccination did not induce an inflammatory response in placental cells. CONCLUSIONS: Our studies demonstrate a significant impact of indirect SARS-CoV-2 infection on gene expression of inner mesenchymal HBCs, with limited effect on lining CTB cells isolated from pregnant subjects infected and recovered from SARS-CoV-2. The pathways associated with these DEGs identify potential targets for therapeutic intervention.


Assuntos
COVID-19 , Placenta , Gravidez , Feminino , Humanos , COVID-19/genética , COVID-19/metabolismo , SARS-CoV-2/genética , Trofoblastos/metabolismo , Transcriptoma , RNA Mensageiro/metabolismo
2.
Front Immunol ; 15: 1339304, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38361952

RESUMO

Melatonin (N-acetyl-5-methoxytryptamine) is an indolamine hormone with many physiological and biological roles. Melatonin is an antioxidant, anti-inflammatory, free radical scavenger, circadian rhythm regulator, and sleep hormone. However, its most popular role is the ability to regulate sleep through the circadian rhythm. Interestingly, recent studies have shown that melatonin is an important and essential hormone during pregnancy, specifically in the placenta. This is primarily due to the placenta's ability to synthesize its own melatonin rather than depending on the pineal gland. During pregnancy, melatonin acts as an antioxidant and anti-inflammatory, which is necessary to ensure a stable environment for both the mother and the fetus. It is an essential antioxidant in the placenta because it reduces oxidative stress by constantly scavenging for free radicals, i.e., maintain the placenta's integrity. In a healthy pregnancy, the maternal immune system is constantly altered to accommodate the needs of the growing fetus, and melatonin acts as a key anti-inflammatory by regulating immune homeostasis during early and late gestation. This literature review aims to identify and summarize melatonin's role as a powerful antioxidant and anti-inflammatory that reduces oxidative stress and inflammation to maintain a favorable homeostatic environment in the placenta throughout gestation.


Assuntos
Melatonina , Gravidez , Feminino , Humanos , Melatonina/farmacologia , Antioxidantes , Placenta , Sequestradores de Radicais Livres , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
3.
J Infect Dis ; 229(2): 473-484, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-37786979

RESUMO

Despite intensive characterization of immune responses after COVID-19 infection and vaccination, research examining protective correlates of vertical transmission in pregnancy are limited. Herein, we profiled humoral and cellular characteristics in pregnant women infected or vaccinated at different trimesters and in their corresponding newborns. We noted a significant correlation between spike S1-specific IgG antibody and its RBD-ACE2 blocking activity (receptor-binding domain-human angiotensin-converting enzyme 2) in maternal and cord plasma (P < .001, R > 0.90). Blocking activity of spike S1-specific IgG was significantly higher in pregnant women infected during the third trimester than the first and second trimesters. Elevated levels of 28 cytokines/chemokines, mainly proinflammatory, were noted in maternal plasma with infection at delivery, while cord plasma with maternal infection 2 weeks before delivery exhibited the emergence of anti-inflammatory cytokines. Our data support vertical transmission of protective SARS-CoV-2-specific antibodies. This vertical antibody transmission and the presence of anti-inflammatory cytokines in cord blood may offset adverse outcomes of inflammation in exposed newborns.


Assuntos
COVID-19 , Complicações Infecciosas na Gravidez , Recém-Nascido , Gravidez , Humanos , Feminino , SARS-CoV-2 , Anticorpos Antivirais , Citocinas , Anti-Inflamatórios
4.
PLoS One ; 18(9): e0290375, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37656705

RESUMO

Staphylococcus aureus (S. aureus) is known to cause human infections and since the late 1990s, community-onset antibiotic resistant infections (methicillin resistant S. aureus (MRSA)) continue to cause significant infections in the United States. Skin and soft tissue infections (SSTIs) still account for the majority of these in the outpatient setting. Machine learning can predict the location-based risks for community-level S. aureus infections. Multi-year (2002-2016) electronic health records of children <19 years old with S. aureus infections were queried for patient level data for demographic, clinical, and laboratory information. Area level data (Block group) was abstracted from U.S. Census data. A machine learning ecological niche model, maximum entropy (MaxEnt), was applied to assess model performance of specific place-based factors (determined a priori) associated with S. aureus infections; analyses were structured to compare methicillin resistant (MRSA) against methicillin sensitive S. aureus (MSSA) infections. Differences in rates of MRSA and MSSA infections were determined by comparing those which occurred in the early phase (2002-2005) and those in the later phase (2006-2016). Multi-level modeling was applied to identify risks factors for S. aureus infections. Among 16,124 unique patients with community-onset MRSA and MSSA, majority occurred in the most densely populated neighborhoods of Atlanta's metropolitan area. MaxEnt model performance showed the training AUC ranged from 0.771 to 0.824, while the testing AUC ranged from 0.769 to 0.839. Population density was the area variable which contributed the most in predicting S. aureus disease (stratified by CO-MRSA and CO-MSSA) across early and late periods. Race contributed more to CO-MRSA prediction models during the early and late periods than for CO-MSSA. Machine learning accurately predicts which densely populated areas are at highest and lowest risk for community-onset S. aureus infections over a 14-year time span.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Humanos , Criança , Adulto Jovem , Adulto , Staphylococcus aureus , Sudeste dos Estados Unidos/epidemiologia , Aprendizado de Máquina , Infecções Estafilocócicas/diagnóstico , Infecções Estafilocócicas/epidemiologia
5.
J Child Fam Stud ; 31(7): 2050-2064, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35221643

RESUMO

Mental illness in adolescence is associated with high-risk sexual behaviors including multiple sex partners, infrequent or inconsistent condom use, and nonuse of contraception. Inpatient psychiatric care represents a promising setting to provide sexual health education. This pilot study investigates the feasibility and acceptability of online sexual health education in this group by assessing usability and impact on short-term psychosocial outcomes. We administered online modules on healthy relationships, pregnancy prevention, condom use, and sexually transmitted infection (STI) prevention to youth. We evaluated outcomes using a single group, pre/post-intervention design. One quality improvement session assessed staff acceptability of the programming. Participants included 51 inpatients (mean age = 15.3; 61% female; 57% Hispanic or Latino; 55% heterosexual). Overall, the program was feasible to administer and highly acceptable to youth (84-89% liked the modules, 98-100% found them easy to use, 96-100% found them credible, 91-98% said information would lead to healthier dating relationships, and 78-87% would refer to a friend). Youth who completed modules demonstrated improvement in several outcomes: attitudes and norms towards violence (p < 0.001), intention to use a method of birth control other than condoms if having sex in the next 3 months (p < 0.001), condom knowledge (p < 0.001), condom use self-efficacy (p < 0.001), condom beliefs (p = 0.04), HIV/STI knowledge (p < 0.001), and perceived susceptibility to STI (p < 0.01). The quality improvement session revealed high acceptability by nursing staff on the unit. This intervention could be useful and efficacious in an inpatient setting and larger studies are warranted to understand its full impact.

6.
Artigo em Inglês | MEDLINE | ID: mdl-36742289

RESUMO

In humans, the hemochorial placenta is a unique temporary organ that forms during pregnancy to support fetal development, gaseous exchange, delivery of nutrition, removal of waste products, and provides immune protection, while maintaining tolerance to the HLA-haploidentical fetus. In this review, we characterize decidual and placental immunity during maternal viral (co)-infection with HIV-1, human cytomegalovirus (HCMV), and Zika virus. We discuss placental immunology, clinical presentation, and epidemiology, before characterizing host susceptibility and cellular tropism, and how the three viruses gain access into specific placental target cells. We describe current knowledge on host-viral interactions with decidual and stromal human placental macrophages or Hofbauer cells, trophoblasts including extra villous trophoblasts, T cells, and decidual natural killer (dNK) cells. These clinically significant viral infections elicit both innate and adaptive immune responses to control replication. However, the three viruses either during mono- or co-infection (HIV-1 and HCMV) escape detection to initiate placental inflammation associated with viral transmission to the developing fetus. Aside from congenital or perinatal infection, other adverse pregnancy outcomes include preterm labor and spontaneous abortion. In addition, maternal HIV-1 and HCMV co-infection are associated with impaired fetal and infant immunity in postnatal life and poor clinical outcomes during childhood in exposed infants, even in the absence of vertical transmission of HIV-1. Given the rapidly expanding numbers of HIV-1-exposed uninfected infants and children globally, further research is urgently needed on neonatal immune programming during maternal mono-and co-infection. This review therefore includes sections on current knowledge gaps that may prompt future research directions. These gaps reflect an emerging but poorly characterized field. Their significance and potential investigation is underscored by the fact that although viral infections result in adverse consequences in both mother and developing fetus/newborn, antiviral and immunomodulatory therapies can improve clinical outcomes in the dyad.

7.
PLoS Pathog ; 17(8): e1009860, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34432853

RESUMO

In 2019, >90% of new HIV infections in infants globally occurred vertically. Studies suggest intrauterine transmission most often occurs in the third trimester; however, there are no mechanistic studies to support these observations. We therefore obtained early/mid-gestation and term placentae from 20 HIV/Hepatitis B/CMV negative women. Isolated primary placental macrophages (Hofbauer cells [HCs]) were exposed to HIV-1BaL and/or interferon (IFN)-α, IFN-ß, IFN-λ1, and RIG-I-like receptor (RLR) agonists. qRT-PCR, FACS, ELISA, Luminex, and Western blot analyses determined expression of activation markers, co-receptors, viral antigen, cytokines, antiviral genes, and host proteins. Early gestation HCs express higher levels of CCR5 and exhibit a more activated phenotype. Despite downregulation of CCR5, term HCs were more susceptible to HIV replication. Early gestation HCs displayed a more activated phenotype than term HCs and HIV exposure lead to the further up-regulation of T-cell co-stimulatory and MHC molecules. Limited HIV replication in early/mid gestation HCs was associated with increased secretion of anti-inflammatory cytokines, chemokines, and a more robust antiviral immune response. In contrast, term HCs were more susceptible to HIV replication, associated with dampening of IFN-induced STAT1 and STAT2 protein activation. Treatment of early/mid gestation and term HCs, with type I IFNs or RLR agonists reduced HIV replication, underscoring the importance of IFN and RLR signaling in inducing an antiviral state. Viral recognition and antiviral immunity in early gestation HCs may prevent in utero HIV infection, whereas diminished antiviral responses at term can facilitate transmission. Defining mechanisms and specific timing of vertical transmission are critical for the development of specific vaccines and antiviral therapeutics to prevent new HIV infections in children globally.


Assuntos
Antivirais/imunologia , Infecções por HIV/prevenção & controle , HIV-1/fisiologia , Imunidade Inata/imunologia , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Placenta/imunologia , Replicação Viral , Adolescente , Feminino , Idade Gestacional , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Macrófagos/imunologia , Macrófagos/virologia , Placenta/virologia , Gravidez
8.
Am J Reprod Immunol ; 85(3): e13358, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33064324

RESUMO

PROBLEM: Prenatal exposure to metabolic dysregulation arising from maternal obesity can have negative health consequences in post-natal life. To date, the specific effects of maternal obesity on fetal immunity at a cellular level have not been well characterized. METHOD OF STUDY: Using cord blood mononuclear cells (CBMCs) and cord plasma (n = 9/group) isolated from infants born to women with a high body mass index (BMI>25kg/m2 ) compared to women with a normal BMI (18-25kg/m2 ), we evaluated differences in immune cell populations using single-cell mass cytometry (CyTOF). CBMCs were matched according to potentially confounding variables, such as maternal and gestational age, ethnicity, smoking status, and gravidity. Statistical results were adjusted for fetal sex. Data were analyzed by viSNE and FlowSOM softwares in Cytobank™ . RESULTS: In newborn CBMCs from women with high BMI, we observed changes in frequency and phenotype of immune cell populations, including significant increases in CD4+ T cells and decreases in myeloid cell populations. IL-12p40 and MDC concentrations were significantly elevated in the high BMI group compared to control. CONCLUSION: This study demonstrates an association between maternal obesity and fetal immunity. Our results warrant following long-term immunologic outcomes and associated clinical risks in children born to women with a high pre-pregnancy BMI.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Sangue Fetal/citologia , Células Mieloides/imunologia , Obesidade Materna/imunologia , Proteínas ADAM/metabolismo , Índice de Massa Corporal , Células Cultivadas , Feminino , Humanos , Recém-Nascido , Subunidade p40 da Interleucina-12/metabolismo , Masculino , Espectrometria de Massas , Fenótipo , Gravidez , Risco , Análise de Célula Única , Proteínas Supressoras de Tumor/metabolismo , Regulação para Cima
9.
J Immunol ; 204(11): 2931-2939, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32321754

RESUMO

During human pregnancy, proinflammatory responses in the placenta can cause severe fetal complications, including growth restriction, preterm birth, and stillbirth. Villitis of unknown etiology (VUE), an inflammatory condition characterized by the infiltration of maternal CD8+ T cells into the placenta, is hypothesized to be secondary to either a tissue rejection response to the haploidentical fetus or from an undiagnosed infection. In this study, we characterized the global TCR ß-chain profile in human T cells isolated from placentae diagnosed with VUE compared with control and infectious villitis-placentae by immunoSEQ. Immunosequencing demonstrated that VUE is driven predominantly by maternal T cell infiltration, which is significantly different from controls and infectious cases; however, these T cell clones show very little overlap between subjects. Mapping TCR clones to common viral epitopes (CMV, EBV, and influenza A) demonstrated that Ag specificity in VUE was equal to controls and significantly lower than CMV-specific clones in infectious villitis. Our data indicate VUE represents an allograft response, not an undetected infection. These observations support the development of screening methods to predict those at risk for VUE and the use of specific immunomodulatory therapies during gestation to improve outcomes in affected fetuses.


Assuntos
Vilosidades Coriônicas/imunologia , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T/genética , Rejeição de Enxerto/imunologia , Inflamação/imunologia , Doenças Placentárias/imunologia , Gravidez/imunologia , Linfócitos T/imunologia , Adulto , Aloenxertos/imunologia , Antígenos Virais/imunologia , Movimento Celular , Estudos de Coortes , Epitopos de Linfócito T/imunologia , Feminino , Feto , Antígenos HLA/imunologia , Humanos , Adulto Jovem
10.
J Immunol ; 204(9): 2380-2391, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32213562

RESUMO

The importance of fetal placental macrophages (Hofbauer cell [HCs]) is underscored by their appearance 18 d postconception and maintenance through term; however, how human HCs evolve during healthy pregnancy and how microenvironment and ontogeny impact phenotype and function remain unknown. In this study, we comprehensively classify human HCs ex vivo, interrogate phenotypic plasticity, and characterize antiviral immune responses through gestation. Activated HCs were abundant in early pregnancy and decreased by term; molecular signatures emphasize inflammatory phenotypes early in gestation. Frequency of HCs with regulatory phenotypes remained high through term. Furthermore, term HCs exhibited blunted responses to stimulation, indicating reduced plasticity. IFN-λ1 is a key placental IFN that appeared less protective than IFN-α, suggesting a potential weakness in antiviral immunity. Ligand-specific responses were temporally regulated: we noted an absence of inflammatory mediators and reduced antiviral gene transcription following RIG-I activation at term despite all HCs producing inflammatory mediators following IFN-γ plus LPS stimulation. Collectively, we demonstrate sequential, evolving immunity as part of the natural history of HCs through gestation.


Assuntos
Imunidade Inata/imunologia , Macrófagos/imunologia , Placenta/imunologia , Adolescente , Antivirais/imunologia , Proteína DEAD-box 58/imunologia , Feminino , Feto/imunologia , Humanos , Inflamação/imunologia , Mediadores da Inflamação/imunologia , Interferon-alfa/imunologia , Ligantes , Fenótipo , Gravidez
11.
Cell Host Microbe ; 24(5): 731-742.e6, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30439342

RESUMO

Zika virus (ZIKV), which emerged in regions endemic to dengue virus (DENV), is vertically transmitted and results in adverse pregnancy outcomes. Antibodies to DENV can cross-react with ZIKV, but whether these antibodies influence ZIKV vertical transmission remains unclear. Here, we find that DENV antibodies increase ZIKV infection of placental macrophages (Hofbauer cells [HCs]) from 10% to over 80% and enhance infection of human placental explants. ZIKV-anti-DENV antibody complexes increase viral binding and entry into HCs but also result in blunted type I interferon, pro-inflammatory cytokine, and antiviral responses. Additionally, ZIKV infection of HCs and human placental explants is enhanced in an immunoglobulin G subclass-dependent manner, and targeting FcRn reduces ZIKV replication in human placental explants. Collectively, these findings support a role for pre-existing DENV antibodies in enhancement of ZIKV infection of select placental cell types and indicate that pre-existing immunity to DENV should be considered when addressing ZIKV vertical transmission.


Assuntos
Anticorpos Antivirais/imunologia , Anticorpos Facilitadores/imunologia , Reações Cruzadas/imunologia , Vírus da Dengue/imunologia , Dengue/imunologia , Macrófagos/imunologia , Placenta/imunologia , Infecção por Zika virus/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Vilosidades Coriônicas , Citocinas/genética , Citocinas/metabolismo , Feminino , Expressão Gênica , Humanos , Imunoglobulina G/imunologia , Transmissão Vertical de Doenças Infecciosas , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Macrófagos/virologia , Gravidez , Internalização do Vírus , Zika virus
12.
J Infect Dis ; 218(9): 1464-1473, 2018 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-29860306

RESUMO

Several co-pathogens that pose threats to the fetus during gestation, including human cytomegalovirus (HCMV), may also contribute to mother-to-child transmission (MTCT) of human immunodeficiency virus type 1 (HIV-1). Within endemic settings, associations between maternal HCMV viral load and increased incidence of MTCT of HIV-1 are documented; however, the mechanisms that promote transmission are poorly characterized. Here we demonstrate that HCMV coinfection enhances susceptibility and viral replication of HIV-1 in placental macrophages (Hofbauer cells) in vitro. Consistent with enhanced viral susceptibility, HCMV exposure upregulates CCR5 and CD80 expression on Hofbauer cells. HCMV also significantly induces type I interferon (IFN), proinflammatory cytokines, and antiviral gene expression. Interestingly, we found that HCMV diminishes type I IFN-mediated phosphorylation of STAT2. Collectively, our data suggest that HCMV-induced activation, local inflammation, and antagonism of type I IFN responses in placental Hofbauer cells promote in utero transmission of HIV-1.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , Infecções por HIV/transmissão , HIV-1/genética , HIV-1/patogenicidade , Placenta/virologia , Replicação Viral/genética , Coinfecção/metabolismo , Coinfecção/virologia , Citocinas/metabolismo , Infecções por Citomegalovirus/metabolismo , Feminino , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Humanos , Transmissão Vertical de Doenças Infecciosas , Inflamação/metabolismo , Inflamação/virologia , Macrófagos/metabolismo , Macrófagos/virologia , Placenta/metabolismo , Gravidez
13.
Pathog Immun ; 2(2): 199-218, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28776046

RESUMO

Zika Virus (ZIKV) is a flavivirus that has been implicated in causing brain deformations, birth defects, and microcephaly in fetuses, and associated with Guillain-Barre syndrome. Mechanisms responsible for transmission of ZIKV across the placenta to the fetus are incompletely understood. Herein, we define key events modulating infection in clinically relevant cells, including primary placental macrophages (human Hofbauer cells; HC), trophoblasts, and neuroblastoma cells. Consistent with previous findings, HC and trophoblasts are permissive to ZIKV infection. Decrease of interferon signaling by Jak ½ inhibition (using ruxolitinib) significantly increased ZIKV replication in HC, trophoblasts, and neuroblasts. Enhanced ZIKV production in ruxolitinib-treated HC was associated with increased expression of HLA-DR and DC-SIGN. Nucleoside analogs blocked ruxolitinib-mediated production of extracellular virus. Although low-level ZIKV infection occurred in untreated HC and trophoblasts, replicating virions were incapable of infecting naive Vero cells. These deficient virions from untreated HC have "thin-coats" suggesting an immature structure. Blocking Jak ½ signaling (with ruxolitinib) restored replication competence as virions produced under these conditions confer cytopathic effects to naive Vero cells. These data demonstrate that Jak-STAT signaling directly impacts the ability of primary placental cells to produce replication-competent virus and is a key determinant in the production of mature virions in clinically relevant cells, including HC and trophoblasts. Design of targeted agents to prevent ZIKV replication in the placenta should consider Jak ½ signaling, the impact of its block on ZIKV infection, and subsequent transmission to the fetus.

14.
Cell Host Microbe ; 20(1): 83-90, 2016 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-27247001

RESUMO

The recent Zika virus (ZIKV) outbreak in Brazil has been directly linked to increased cases of microcephaly in newborns. Current evidence indicates that ZIKV is transmitted vertically from mother to fetus. However, the mechanism of intrauterine transmission and the cell types involved remain unknown. We demonstrate that the contemporary ZIKV strain PRVABC59 (PR 2015) infects and replicates in primary human placental macrophages, called Hofbauer cells, and to a lesser extent in cytotrophoblasts, isolated from villous tissue of full-term placentae. Viral replication coincides with induction of type I interferon (IFN), pro-inflammatory cytokines, and antiviral gene expression, but with minimal cell death. Our results suggest a mechanism for intrauterine transmission in which ZIKV gains access to the fetal compartment by directly infecting placental cells and disrupting the placental barrier.


Assuntos
Macrófagos/virologia , Placenta/citologia , Tropismo Viral , Replicação Viral , Zika virus/fisiologia , Morte Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Humanos , Gravidez , Trofoblastos/virologia
15.
Curr Opin Infect Dis ; 29(3): 248-55, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27027245

RESUMO

PURPOSE OF REVIEW: Mother-to-child transmission (MTCT) of HIV-1 remains a significant global health concern despite implementation of maternal combination antiretroviral therapy for treatment as prevention to offset transmission. The risk of in-utero HIV-1 transmission in the absence of interventions is ∼7%. This low rate of transmission points to innate and adaptive mechanisms to restrict lentiviral infection within the placenta. RECENT FINDINGS: Placental macrophages (Hofbauer cells) are key mediators in in-utero transmission of HIV-1. Hofbauer cells constitutively express elevated concentrations of regulatory cytokines, which inhibit HIV-1 replication in vitro, and possess intrinsic antiviral properties. Hofbauer cells sequester HIV-1 in intracellular compartments that can be accessed by HIV-1-specific antibodies and may occur in vivo to offset MTCT. Intriguingly, studies have reported strong associations between maternal human cytomegalovirus (HCMV) viremia and MTCT of HIV-1. HCMV infection at the placenta promotes inflammation, chronic villitis, and trophoblast damage, providing potential HIV-1 access into CD4CCR5 target cells. The placenta exhibits a variety of mechanisms to limit HIV-1 replication, yet viral-induced activation with maternal HCMV may override this protection to facilitate in-utero transmission of HIV-1. SUMMARY: Understanding immune correlates of protection or transmission at the placenta during on-going HIV-1 exposure may contribute to understanding HIV pathogenesis and the development of effective immunotherapies.


Assuntos
Infecções por HIV , HIV-1/imunologia , Transmissão Vertical de Doenças Infecciosas , Placenta , Feminino , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Humanos , Placenta/citologia , Placenta/imunologia , Gravidez
16.
J Int AIDS Soc ; 18: 19385, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25623930

RESUMO

INTRODUCTION: Within monocyte-derived macrophages, HIV-1 accumulates in intracellular virus-containing compartments (VCCs) that are inaccessible to the external environment, which implicate these cells as latently infected HIV-1 reservoirs. During mother-to-child transmission of HIV-1, human placental macrophages (Hofbauer cells (HCs)) are viral targets, and have been shown to be infected in vivo and sustain low levels of viral replication in vitro; however, the risk of in utero transmission is less than 7%. The role of these primary macrophages as viral reservoirs is largely undefined. The objective of this study is to define potential sites of viral assembly, accumulation and neutralization in HCs given the pivotal role of the placenta in preventing HIV-1 infection in the mother-infant dyad. METHODS: Term placentae from 20 HIV-1 seronegative women were obtained following caesarian section. VCCs were evaluated by 3D confocal and electron microscopy. Colocalization R values (Pearson's correlation) were quantified with colocalization module of Volocity 5.2.1. Replication kinetics and neutralization studies were evaluated using p24 ELISA. RESULTS: We demonstrate that primary HCs assemble and sequester HIV-1(BaL) in intracellular VCCs, which are enriched in endosomal/lysosomal markers, including CD9, CD81, CD63 and LAMP-1. Following infection, we observed HIV-1 accumulation in potentially acidic compartments, which stained intensely with Lysotracker-Red. Remarkably, these compartments are readily accessible via the cell surface and can be targeted by exogenously applied small molecules and HIV-1-specific broadly neutralizing antibodies. In addition, broadly neutralizing antibodies (4E10 and VRC01) limited viral replication by HIV-1-infected HCs, which may be mediated by FcγRI. CONCLUSIONS: These findings suggest that placental HCs possess intrinsic adaptations facilitating unique sequestration of HIV-1, and may serve as a protective viral reservoir to permit viral neutralization and/or antiretroviral drug entry in utero.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Anti-HIV/imunologia , HIV-1/fisiologia , Macrófagos/virologia , Placenta/virologia , Tetraspaninas/análise , Montagem de Vírus , Feminino , Humanos , Placenta/citologia , Gravidez , Replicação Viral
17.
J Infect Dis ; 211(2): 187-96, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25081935

RESUMO

Administration of combination antiretroviral therapy to human immunodeficiency virus type 1 (HIV-1)-infected pregnant women significantly reduces vertical transmission. In contrast, maternal co-opportunistic infection with primary or reactivated cytomegalovirus (CMV) or other pathogens may facilitate in utero transmission of HIV-1 by activation of cord blood mononuclear cells (CBMCs). Here we examine the targets and mechanisms that affect fetal susceptibility to HIV-1 in utero. Using flow cytometry, we demonstrate that the fraction of CD4(+)CD45RO(+) and CD4(+)CCR5(+) CBMCs is minimal, which may account for the low level of in utero HIV-1 transmission. Unstimulated CD4(+) CBMCs that lack CCR5/CD45RO showed reduced levels of HIV-1 infection. However, upon in vitro stimulation with CMV, CBMCs undergo increased proliferation to upregulate the fraction of T central memory cells and expression of CCR5, which enhances susceptibility to HIV-1 infection in vitro. These data suggest that activation induced by CMV in vivo may alter CCR5 expression in CD4(+) T central memory cells to promote in utero transmission of HIV-1.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citomegalovirus/imunologia , Sangue Fetal/imunologia , Infecções por HIV/transmissão , HIV-1/fisiologia , Receptores CCR5/análise , Adulto , Linfócitos T CD4-Positivos/química , Proliferação de Células , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Gravidez
18.
J Child Sex Abus ; 23(5): 504-18, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24830829

RESUMO

Research suggests parents of sexually abused children may experience negative perceptions of themselves and their parenting abilities following the victimization of their children, which may influence the recovery process in treatment for these families. This study assessed perceived self-efficacy and parenting satisfaction among female caregivers of sexually abused children and female caregivers whose children were not victimized. Results indicated that caregivers of child victims had significantly lower levels of perceived parenting efficacy than their counterparts. There were no significant differences found between groups with regard to parenting satisfaction. The results highlight the importance of evaluating perceived parenting competence in the treatment of nonoffending caregivers in order to facilitate optimal treatment of the child victim and all members of the family system.


Assuntos
Cuidadores/psicologia , Abuso Sexual na Infância/psicologia , Poder Familiar/psicologia , Satisfação Pessoal , Autoimagem , Autoeficácia , Adulto , Criança , Feminino , Humanos , Pessoa de Meia-Idade , Relações Pais-Filho , Adulto Jovem
19.
Retrovirology ; 9: 101, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23217137

RESUMO

BACKGROUND: Despite readily detectable levels of the HIV-1 (co)-receptors CD4, CCR5 and DC-SIGN on placental macrophages (Hofbauer Cells [HCs]), the rate of HIV-1 infection in utero in the absence of interventions is only 7% of exposed infants. Here, we examine the replication kinetics of human HCs to the primary isolate HIV-1BaL. We also determined the infectivity of HIV-1-exposed HCs by co-culturing with isolated cord and peripheral blood mononuclear cells [CBMCs, PBMCs]. To understand the limiting nature of HCs to HIV-1 replication, we examined the effect of endogenously secreted cytokines on replication kinetics. RESULTS: HCs have reduced ability to replicate HIV-1 in vitro (p < 0.01) and to transmit virus to CBMCs and PBMCs (p < 0.001 for both) compared to standard infections of MDMs. HCs were shown to release HIV-1 particles at levels comparable to MDMs, however exhibit significant decreases in viral transcription (gag and env), which may account for lower levels of HIV-1 replication. Un-stimulated HCs constitutively express significantly higher levels of regulatory cytokines, IL-10 and TGF-ß, compared to MDMs (p < 0.01), which may contribute to immunoregulatory predominance at the placenta and possibly account for down-regulation of HIV-1 replication and infectivity by HCs. We further demonstrate that these regulatory cytokines inhibit HIV-1 replication within HCs in vitro. CONCLUSION: HCs have reduced ability to replicate and disseminate R5-tropic HIV-1BaLin vitro and potentially offset mother to child transmission (MTCT) of HIV-1 by the induction of immunoregulatory cytokines. Despite the potential for migration and infectivity, HCs are not present in the neighboring fetal circulation. These results implicate HCs as important mediators of protection at the feto-maternal interface during ongoing HIV-1 exposure.


Assuntos
Síndrome da Imunodeficiência Adquirida/transmissão , Citocinas/biossíntese , HIV-1/fisiologia , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Macrófagos/fisiologia , Placenta/virologia , Replicação Viral , Membrana Celular/virologia , Quimiocina CCL3/fisiologia , Quimiocina CCL4/fisiologia , Feminino , Humanos , Placenta/citologia , Gravidez , Receptores CCR5/fisiologia , Montagem de Vírus
20.
World J Surg Oncol ; 9: 46, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21539750

RESUMO

BACKGROUND: Chemotherapy heavily relies on apoptosis to kill breast cancer (BrCa) cells. Many breast tumors respond to chemotherapy, but cells that survive this initial response gain resistance to subsequent treatments. This leads to aggressive cell variants with an enhanced ability to migrate, invade and survive at secondary sites. Metastasis and chemoresistance are responsible for most cancer-related deaths; hence, therapies designed to minimize both are greatly needed. We have recently shown that CCR9-CCL25 interactions promote BrCa cell migration and invasion, while others have shown that this axis play important role in T cell survival. In this study we have shown potential role of CCR9-CCL25 axis in breast cancer cell survival and therapeutic efficacy of cisplatin. METHODS: Bromodeoxyuridine (BrdU) incorporation, Vybrant apoptosis and TUNEL assays were performed to ascertain the role of CCR9-CCL25 axis in cisplatin-induced apoptosis of BrCa cells. Fast Activated Cell-based ELISA (FACE) assay was used to quantify In situ activation of PI3Kp85, AktSer473, GSK-3ßSer9 and FKHRThr24 in breast cancer cells with or without cisplatin treatment in presence or absence of CCL25. RESULTS: CCR9-CCL25 axis provides survival advantage to BrCa cells and inhibits cisplatin-induced apoptosis in a PI3K-dependent and focal adhesion kinase (FAK)-independent fashion. Furthermore, CCR9-CCL25 axis activates cell-survival signals through Akt and subsequent glycogen synthase kinase-3 beta (GSK-3ß) and forkhead in human rhabdomyosarcoma (FKHR) inactivation. These results show that CCR9-CCL25 axis play important role in BrCa cell survival and low chemotherapeutic efficacy of cisplatin primarily through PI3K/Akt dependent fashion.


Assuntos
Antineoplásicos/farmacologia , Quimiocinas CC/metabolismo , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Receptores CCR/metabolismo , Apoptose , Neoplasias da Mama , Movimento Celular , Sobrevivência Celular , Quinase 1 de Adesão Focal/metabolismo , Humanos , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA