Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 392
Filtrar
1.
Vet Microbiol ; 295: 110154, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38959808

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is one of the costliest diseases to pork producers worldwide. We tested samples from the pregnant gilt model (PGM) to better understand the fetal response to in-utero PRRS virus (PRRSV) infection. Our goal was to identify critical tissues and genes associated with fetal resilience or susceptibility. Pregnant gilts (N=22) were infected with PRRSV on day 86 of gestation. At 21 days post maternal infection, the gilts and fetuses were euthanized, and fetal tissues collected. Fetuses were characterized for PRRS viral load in fetal serum and thymus, and preservation status (viable or meconium stained: VIA or MEC). Fetuses (N=10 per group) were compared: uninfected (UNIF; <1 log/µL PRRSV RNA), resilient (HV_VIA, >5 log virus/µL but viable), and susceptible (HV_MEC, >5 log virus/µL with MEC). Gene expression in fetal heart, kidney, and liver was investigated using NanoString transcriptomics. Gene categories investigated were hypothesized to be involved in fetal response to PRRSV infection: renin- angiotensin-aldosterone, inflammatory, transporter and metabolic systems. Following PRRSV infection, CCL5 increased expression in heart and kidney, and ACE2 decreased expression in kidney, each associated with fetal PRRS susceptibility. Liver revealed the most significant differential gene expression: CXCL10 decreased and IL10 increased indicative of immune suppression. Increased liver gene expression indicated potential associations with fetal PRRS susceptibility on several systems including blood pressure regulation (AGTR1), energy metabolism (SLC16A1 and SLC16A7), tissue specific responses (KL) and growth modulation (TGFB1). Overall, analyses of non-lymphoid tissues provided clues to mechanisms of fetal compromise following maternal PRRSV infection.


Asunto(s)
Resistencia a la Enfermedad , Feto , Síndrome Respiratorio y de la Reproducción Porcina , Transcriptoma , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Virus del Síndrome Respiratorio y Reproductivo Porcino/inmunología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Embarazo , Animales , Porcinos , Femenino , Feto/inmunología , Feto/virología , Regulación de la Expresión Génica/inmunología , Miocardio/inmunología , Hígado/inmunología , Susceptibilidad a Enfermedades/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/veterinaria , Riñón/inmunología
2.
BMC Vet Res ; 20(1): 255, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38867209

RESUMEN

BACKGROUND: Porcine reproductive and respiratory syndrome virus 2 (PRRSV-2) infection during late gestation substantially lowers fetal viability and survival. In a previous genome-wide association study, a single nucleotide polymorphism on chromosome 7 was significantly associated with probability of fetuses being viable in response to maternal PRRSV-2 infection at 21 days post maternal inoculation. The iodothyronine deiodinase 2 (DIO2) gene, located ~ 14 Kilobase downstream of this SNP, was selected as a priority candidate related to fetal susceptibility following maternal PRRSV-2 infection. Our objectives were to identify mutation(s) within the porcine DIO2 gene and to determine if they were associated with fetal outcomes after PRRSV-2 challenge. Sequencing of the DIO2, genotyping identified variants, and association of DIO2 genotypes with fetal phenotypes including DIO2 mRNA levels, viability, survival, viral loads, cortisol and thyroid hormone levels, and growth measurements were conducted. RESULTS: A missense variant (p.Asn91Ser) was identified in the parental populations from two independent PRRSV-2 challenge trials. This variant was further genotyped to determine association with fetal PRRS outcomes. DIO2 mRNA levels in fetal heart and kidney differed by the genotypes of Asn91Ser substitution with significantly greater DIO2 mRNA expression in heterozygotes compared with wild-type homozygotes (P < 0.001 for heart, P = 0.002 for kidney). While Asn91Ser did not significantly alter fetal viability and growth measurements, interaction effects of the variant with fetal sex or trial were identified for fetal viability or crown rump length, respectively. However, this mutation was not related to dysregulation of the hypothalamic-pituitary-adrenal and thyroid axis, indicated by no differences in circulating cortisol, T4, and T3 levels in fetuses of the opposing genotypes following PRRSV-2 infection. CONCLUSIONS: The present study suggests that a complex relationship among DIO2 genotype, DIO2 expression, fetal sex, and fetal viability may exist during the course of fetal PRRSV infection. Our study also proposes the increase in cortisol levels, indicative of fetal stress response, may lead to fetal complications, such as fetal compromise, fetal death, or premature farrowing, during PRRSV infection.


Asunto(s)
Yoduro Peroxidasa , Mutación Missense , Síndrome Respiratorio y de la Reproducción Porcina , Virus del Síndrome Respiratorio y Reproductivo Porcino , Animales , Síndrome Respiratorio y de la Reproducción Porcina/genética , Síndrome Respiratorio y de la Reproducción Porcina/virología , Femenino , Porcinos , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Embarazo , Yodotironina Deyodinasa Tipo II , Genotipo , Feto/virología
3.
Semin Perinatol ; 48(4): 151919, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38897829

RESUMEN

Pregnant people and their fetuses are vulnerable to adverse health outcomes from coronavirus 2019 disease (COVID-19) due to infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). COVID-19 has been associated with higher rates of maternal mortality, preterm birth, and stillbirth. While SARS-CoV-2 infection of the placenta and vertical transmission is rare, this may be due to the typically longer time interval between maternal infection and testing of the placenta and neonate. Placental injury is evident in cases of SARS-CoV-2-associated stillbirth with massive perivillous fibrin deposition, chronic histiocytic intervillositis, and trophoblast necrosis. Maternal COVID-19 can also polarize fetal immunity, which may have long-term effects on neurodevelopment. Although the COVID-19 pandemic continues to evolve, the impact of emerging SARS-CoV-2 variants on placental and perinatal injury/mortality remains concerning for maternal and perinatal health. Here, we highlight the impact of COVID-19 on the placenta and fetus and remaining knowledge gaps.


Asunto(s)
COVID-19 , Transmisión Vertical de Enfermedad Infecciosa , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Humanos , Embarazo , COVID-19/transmisión , Femenino , Placenta/virología , Recién Nacido , Mortinato , Feto/virología , Enfermedades Placentarias/virología , Nacimiento Prematuro
4.
Viruses ; 16(6)2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38932183

RESUMEN

Congenital cytomegalovirus (cCMV) infection poses significant risks to fetal development, particularly affecting the nervous system. This study reports a fetal autopsy case, examining cCMV infection and focusing on CMV DNA measurements in various fetal organs before formalin fixation, a novel approach for comprehensive CMV DNA evaluations in fetal organs affected by cCMV. A 20-week-old male fetus was diagnosed with cCMV following the detection of CMV DNA in ascites obtained via abdominocentesis in utero. After the termination of pregnancy, multiple organs of the fetus, including the cerebrum, thyroid gland, heart, lungs, liver, spleen, kidneys, and adrenal glands, were extracted and examined for CMV DNA loads using a real-time polymerase chain reaction. Histopathological examination involved hematoxylin-eosin and CMV-specific immunostaining. A correlation was found between CMV DNA loads and pathology, with higher CMV-infected cell numbers observed in organs positively identified with both staining methods, exhibiting CMV DNA levels of ≥1.0 × 104 copies/mL, compared to those detected solely by CMV-specific immunostaining, where CMV DNA levels ranged from 1.0 × 103 to 1.0 × 104 copies/mL. These results highlight a quantifiable relationship between the organ infection extent and CMV DNA concentration, providing insights into cCMV pathogenesis and potentially informing future diagnostic and therapeutic strategies for cCMV infection.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , ADN Viral , Feto , Carga Viral , Infecciones por Citomegalovirus/congénito , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/diagnóstico , Humanos , Citomegalovirus/genética , Citomegalovirus/aislamiento & purificación , ADN Viral/genética , Masculino , Femenino , Feto/virología , Embarazo , Adulto , Autopsia , Complicaciones Infecciosas del Embarazo/virología
5.
Arch Virol ; 169(6): 133, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38829449

RESUMEN

Akabane virus (AKAV), Aino virus, Peaton virus, Sathuperi virus, and Shamonda virus are arthropod-borne viruses belonging to the order Elliovirales, family Peribunyaviridae, genus Orthobunyavirus. These viruses cause or may cause congenital malformations in ruminants, including hydranencephaly, poliomyelitis, and arthrogryposis, although their pathogenicity may vary among field cases. AKAV may cause relatively severe congenital lesions such as hydranencephaly in calves. Furthermore, strains of AKAV genogroups I and II exhibit different disease courses. Genogroup I strains predominantly cause postnatal viral encephalomyelitis, while genogroup II strains are primarily detected in cases of congenital malformation. However, the biological properties of AKAV and other orthobunyaviruses are insufficiently investigated in hosts in the field and in vitro. Here, we used an immortalized bovine brain cell line (FBBC-1) to investigate viral replication efficiency, cytopathogenicity, and host innate immune responses. AKAV genogroup II and Shamonda virus replicated to higher titers in FBBC-1 cells compared with the other viruses, and only AKAV caused cytopathic effects. These results may be associated with the severe congenital lesions in the brain caused by AKAV genogroup II. AKAV genogroup II strains replicated to higher titers in FBBC-1 cells than AKAV genogroup I strains, suggesting that genogroup II strains replicated more efficiently in fetal brain cells, accounting for the detection of the latter strains mainly in fetal infection cases. Therefore, FBBC-1 cells may serve as a valuable tool for investigating the virulence and tropism of the orthobunyaviruses for bovine neonatal brain tissues in vitro.


Asunto(s)
Encéfalo , Infecciones por Bunyaviridae , Orthobunyavirus , Replicación Viral , Animales , Bovinos , Orthobunyavirus/patogenicidad , Orthobunyavirus/genética , Orthobunyavirus/fisiología , Orthobunyavirus/clasificación , Encéfalo/virología , Encéfalo/patología , Línea Celular , Infecciones por Bunyaviridae/virología , Infecciones por Bunyaviridae/veterinaria , Infecciones por Bunyaviridae/patología , Enfermedades de los Bovinos/virología , Feto/virología , Efecto Citopatogénico Viral , Inmunidad Innata
6.
Sci Rep ; 14(1): 11422, 2024 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-38763951

RESUMEN

Our center has observed a substantial increase in the detection rate of fetal left-right(LR) asymmetry disorders between March and May 2023. This finding has raised concerns because these pregnant women experienced the peak outbreak of SARS-CoV-2 in China during their first trimester. To explore the relationship between maternal SARS-CoV-2 infection and fetal LR asymmetry disorders. A retrospective collection of clinical and ultrasound data diagnosed as fetal LR asymmetry disorders was conducted from January 2018 to December 2023. The case-control study involved fetuses with LR asymmetry disorders and normal fetuses in a 1:1 ratio. We evaluated and compared the clinical and fetal ultrasound findings in pregnant women with SARS-CoV-2 infection and pregnant women without infection. The Student t-test was utilized to compare continuous variables, while the chi-squared test was employed for univariable analyses. The incidence rate of LR asymmetry disorders from 2018 to 2023 was as follows: 0.17‰, 0.63‰, 0.61‰, 0.57‰, 0.59‰, and 3.24‰, respectively. A total of 30 fetuses with LR asymmetry disorders and 30 normal fetuses were included. This case-control study found that SARS-CoV-2 infection (96.67% vs 3.33%, P = .026) and infection during the first trimester (96.55% vs 3.45%, P = .008) were identified as risk factors. The odds ratio values were 10.545 (95% CI 1.227, 90.662) and 13.067 (95% CI 1.467, 116.419) respectively. In cases of SARS-CoV-2 infection in the first trimester, the majority of infections (88.1%, 37/42) occurred between 5 and 6 weeks of gestation. We found that 43.7% (66/151) of fetuses with LR asymmetry disorder had associated malformations, 90.9% (60/66) exhibited cardiac malformations. SARS-CoV-2 infection during the first trimester significantly increases the risk of fetal LR asymmetry disorders, particularly when the infection occurs between 5 and 6 gestation weeks. The most common associated malformation is heart malformation.


Asunto(s)
COVID-19 , Complicaciones Infecciosas del Embarazo , Primer Trimestre del Embarazo , SARS-CoV-2 , Humanos , Femenino , Embarazo , COVID-19/epidemiología , COVID-19/complicaciones , Complicaciones Infecciosas del Embarazo/epidemiología , Adulto , Estudios Retrospectivos , Estudios de Casos y Controles , China/epidemiología , Ultrasonografía Prenatal , Factores de Riesgo , Feto/virología , Enfermedades Fetales/epidemiología , Enfermedades Fetales/virología
7.
Braz J Microbiol ; 55(2): 1949-1959, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38696038

RESUMEN

Ovine gammaherpesvirus 2 (OvGHV2) produces sheep-associated malignant catarrhal fever (SA-MCF), a frequently lethal, lymphoproliferative disease that is characterized by widespread vascular lesions. Most studies that evaluated the viral load in tissues of animals with SA-MCF were done in the Northern Hemisphere, with scant information from the Southern part of the globe. This study investigated the viral load of OvGHV2 in the tissues of cattle and an underdeveloped fetus with SA-MCF from three distinct biomes of Brazil. All animals had clinical and histopathological manifestations consistent with SA-MCF. Molecular testing identified the OvGHV2 tegument protein and glycoprotein B genes in the tissues of all animals and the fetus. Viral quantification based on the DNA polymerase gene detected elevated loads of OvGHV2 in tissues with histopathological evidence of SA-MCF and organs with unknown histological data, except for the tissues of the fetus, where the viral load was comparatively reduced. The viral loads detected in multiple organs of cattle from this study with SA-MCF are consistent with those identified in different animal species from the USA and Europe. The detection of a low viral load of OvGHV2 in fetal tissue confirmed transplacental dissemination since elevated viral loads were detected in multiple tissues of the cow with SA-MCF. Furthermore, the elevated viral loads detected in the pulmonary tissues of cattle with interstitial pneumonia indicate that OvGHV2 is an inductor of pulmonary disease in cattle.


Asunto(s)
Gammaherpesvirinae , Fiebre Catarral Maligna , Carga Viral , Animales , Fiebre Catarral Maligna/virología , Fiebre Catarral Maligna/patología , Gammaherpesvirinae/aislamiento & purificación , Gammaherpesvirinae/genética , Bovinos , Brasil , Ovinos , Femenino , Enfermedades de las Ovejas/virología , Enfermedades de las Ovejas/patología , ADN Viral/genética , Enfermedades de los Bovinos/virología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Feto/virología
8.
Viruses ; 16(5)2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38793603

RESUMEN

Bovine viral diarrhea virus (BVDV) infections cause USD 1.5-2 billion in losses annually. Maternal BVDV after 150 days of gestation causes transient fetal infection (TI) in which the fetal immune response clears the virus. The impact of fetal TI BVDV infections on postnatal growth and white blood cell (WBC) methylome as an index of epigenetic modifications was examined by inoculating pregnant heifers with noncytopathic type 2 BVDV or media (sham-inoculated controls) on Day 175 of gestation to generate TI (n = 11) and control heifer calves (n = 12). Fetal infection in TI calves was confirmed by virus-neutralizing antibody titers at birth and control calves were seronegative. Both control and TI calves were negative for BVDV RNA in WBCs by RT-PCR. The mean weight of the TI calves was less than that of the controls (p < 0.05). DNA methyl seq analysis of WBC DNA demonstrated 2349 differentially methylated cytosines (p ≤ 0.05) including 1277 hypomethylated cytosines, 1072 hypermethylated cytosines, 84 differentially methylated regions based on CpGs in promoters, and 89 DMRs in islands of TI WBC DNA compared to controls. Fetal BVDV infection during late gestation resulted in epigenomic modifications predicted to affect fetal development and immune pathways, suggesting potential consequences for postnatal growth and health of TI cattle.


Asunto(s)
Diarrea Mucosa Bovina Viral , Metilación de ADN , Virus de la Diarrea Viral Bovina , Epigénesis Genética , Leucocitos , Animales , Bovinos , Diarrea Mucosa Bovina Viral/virología , Diarrea Mucosa Bovina Viral/genética , Femenino , Embarazo , Leucocitos/virología , Virus de la Diarrea Viral Bovina/genética , Anticuerpos Antivirales/sangre , Enfermedades Fetales/virología , Enfermedades Fetales/veterinaria , Enfermedades Fetales/genética , Virus de la Diarrea Viral Bovina Tipo 2/genética , Feto/virología
9.
Cell Rep Med ; 5(5): 101515, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38631348

RESUMEN

During pregnancy, germline development is vital for maintaining the continuation of species. Recent studies have shown increased pregnancy risks in COVID-19 patients at the perinatal stage. However, the potential consequence of infection for reproductive quality in developing fetuses remains unclear. Here, we analyze the transcriptome and DNA methylome of the fetal germline following maternal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We find that infection at early gestational age, a critical period of human primordial germ cell specification and epigenetic reprogramming, trivially affects fetal germ cell (FGC) development. Additionally, FGC-niche communications are not compromised by maternal infection. Strikingly, both general and SARS-CoV-2-specific immune pathways are greatly activated in gonadal niche cells to protect FGCs from maternal infection. Notably, there occurs an "in advance" development tendency in FGCs after maternal infection. Our study provides insights into the impacts of maternal SARS-CoV-2 infection on fetal germline development and serves as potential clinical guidance for future pandemics.


Asunto(s)
COVID-19 , Feto , Células Germinativas , SARS-CoV-2 , Humanos , Femenino , COVID-19/virología , COVID-19/inmunología , COVID-19/patología , Embarazo , Células Germinativas/virología , Feto/virología , Complicaciones Infecciosas del Embarazo/virología , Complicaciones Infecciosas del Embarazo/patología , Gónadas/virología , Transcriptoma/genética , Masculino , Metilación de ADN/genética , Epigénesis Genética
10.
Viruses ; 13(9)2021 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-34578389

RESUMEN

Zika virus (ZIKV) infection during pregnancy causes a wide spectrum of congenital abnormalities and postnatal developmental sequelae such as fetal loss, intrauterine growth restriction (IUGR), microcephaly, or motor and neurodevelopmental disorders. Here, we investigated whether a mouse pregnancy model recapitulated a wide range of symptoms after congenital ZIKV infection, and whether the embryonic age of congenital infection changed the fetal or postnatal outcomes. Infection with ZIKV strain PRVABC59 from embryonic day 6.5 (E6.5) to E8.5, corresponding to the mid-first trimester in humans, caused fetal death, fetal resorption, or severe IUGR, whereas infection from E9.5 to E14.5, corresponding to the late-first to second trimester in humans, caused stillbirth, neonatal death, microcephaly, and postnatal growth deficiency. Furthermore, 4-week-old offspring born to dams infected at E12.5 showed abnormalities in neuropsychiatric state, motor behavior, autonomic function, or reflex and sensory function. Thus, our model recapitulated the multiple symptoms seen in human cases, and the embryonic age of congenital infection was one of the determinant factors of offspring outcomes in mice. Furthermore, maternal neutralizing antibodies protected the offspring from neonatal death after congenital infection at E9.5, suggesting that neonatal death in our model could serve as criteria for screening of vaccine candidates.


Asunto(s)
Feto/virología , Microcefalia/virología , Malformaciones del Sistema Nervioso/virología , Infección por el Virus Zika/congénito , Virus Zika/patogenicidad , Animales , Modelos Animales de Enfermedad , Embrión de Mamíferos/virología , Femenino , Ratones , Ratones Endogámicos C57BL , Embarazo
11.
Viruses ; 13(9)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34578408

RESUMEN

The host's immune status may affect virus evolution. Little is known about how developing fetal and placental immune milieus affect virus heterogeneity. This knowledge will help us better understand intra-host virus evolution and how new virus variants emerge. The goal of our study was to find out whether the isolated in utero environment-an environment with specialized placental immunity and developing fetal immunity-supports the emergence of RNA and DNA virus variants. We used well-established porcine models for isolated Zika virus (RNA virus) and porcine circovirus 2 (DNA virus) fetal infections. We found that the isolated in utero environment was conducive to the emergence of RNA and DNA virus variants. Next-generation sequencing of nearly whole virus genomes and validated bioinformatics pipelines identified both unique and convergent single nucleotide variations in virus genomes isolated from different fetuses. Zika virus and PCV2 in utero evolution also resulted in single nucleotide variations previously reported in the human and porcine field samples. These findings should encourage further studies on virus evolution in placenta and fetuses, to better understand how virus variants emerge and how in utero viral evolution affects congenital virus transmission and pathogenicity.


Asunto(s)
Circovirus/genética , Evolución Molecular Dirigida , Variación Genética , Placenta/virología , Útero/virología , Virus Zika/genética , Animales , Microambiente Celular , Chlorocebus aethiops , Circovirus/fisiología , Femenino , Feto/virología , Heterogeneidad Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Placenta/inmunología , Embarazo , Porcinos , Útero/inmunología , Células Vero , Carga Viral , Virus Zika/fisiología
12.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166248, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34461257

RESUMEN

The COVID-19 pandemic has infected nearly 178 million people and claimed the lives of over 3.8 million in less than 15 months. This has prompted a flurry of research studies into the mechanisms and effects of SARS-CoV-2 viral infection in humans. However, studies examining the effects of COVID-19 in pregnant women, their placentae and their babies remain limited. Furthermore, reports of safety and efficacy of vaccines for SARS-CoV-2 in pregnancy are limited. This review concisely summarises the case studies and research on COVID-19 in pregnancy, to date. It also reviews the mechanism of infection with SARS-CoV-2, and its reliance and effects upon the renin-angiotensin-aldosterone system. Overall, the data suggest that infection during pregnancy can be dangerous at any time, but this risk to both the mother and fetus, as well as placental damage, increases during the third trimester. The possibility of vertical transmission, which is explored in this review, remains contentious. However, maternal infection with SARS-CoV-2 can increase risk of miscarriage, preterm birth and stillbirth, which is likely due to damage to the placenta.


Asunto(s)
COVID-19/metabolismo , Feto/inmunología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Complicaciones Infecciosas del Embarazo/virología , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Femenino , Feto/virología , Humanos , Recién Nacido , Pandemias , Placenta/metabolismo , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/prevención & control , SARS-CoV-2/aislamiento & purificación
13.
Cell Rep Med ; 2(7): 100315, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34337555

RESUMEN

Study of evolution and selection pressure on HIV-1 in fetuses will lead to a better understanding of the role of immune responses in shaping virus evolution and vertical transmission. Detailed genetic analyses of HIV-1 env gene from 12 in utero transmission pairs show that most infections (67%) occur within 2 months of childbirth. In addition, the env sequences from long-term-infected fetuses are highly divergent and form separate phylogenetic lineages from their cognate maternal viruses. Host-selection sites unique to neonate viruses are identified in regions frequently targeted by neutralizing antibodies and T cell immune responses. Identification of unique selection sites in the env gene of fetal viruses indicates that the immune system in fetuses is capable of exerting selection pressure on viral evolution. Studying selection and evolution of HIV-1 or other viruses in fetuses can be an alternative approach to investigate adaptive immunity in fetuses.


Asunto(s)
Evolución Biológica , Feto/inmunología , Feto/virología , VIH-1/fisiología , Transmisión Vertical de Enfermedad Infecciosa , Madres , Adolescente , Adulto , Secuencia de Bases , Línea Celular , Femenino , Variación Genética , Infecciones por VIH/sangre , Infecciones por VIH/virología , Interacciones Huésped-Patógeno/inmunología , Humanos , Recién Nacido , Pruebas de Neutralización , Motivos de Nucleótidos/genética , Filogenia , Placenta/metabolismo , Embarazo , Adulto Joven , Productos del Gen env del Virus de la Inmunodeficiencia Humana
14.
J Gen Virol ; 102(8)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34410903

RESUMEN

An infectious agent's pathogenic and transmission potential is heavily influenced by early events during the asymptomatic or subclinical phase of disease. During this phase, the presence of infectious agent may be relatively low. An important example of this is Zika virus (ZIKV), which can cross the placenta and infect the foetus, even in mothers with subclinical infections. These subclinical infections represent roughly 80 % of all human infections. Initial ZIKV pathogenesis studies were performed in type I interferon receptor (IFNAR) knockout mice. Blunting the interferon response resulted in robust infectivity, and increased the utility of mice to model ZIKV infections. However, due to the removal of the interferon response, the use of these models impedes full characterization of immune responses to ZIKV-related pathologies. Moreover, IFNAR-deficient models represent severe disease whereas less is known regarding subclinical infections. Investigation of the anti-viral immune response elicited at the maternal-foetal interface is critical to fully understand mechanisms involved in foetal infection, foetal development, and disease processes recognized to occur during subclinical maternal infections. Thus, immunocompetent experimental models that recapitulate natural infections are needed. We have established subclinical intravaginal ZIKV infections in mice and guinea pigs. We found that these infections resulted in: the presence of both ZIKV RNA transcripts and infectious virus in maternal and placental tissues, establishment of foetal infections and ZIKV-mediated CXCL10 expression. These models will aid in discerning the mechanisms of subclinical ZIKV mother-to-offspring transmission, and by extension can be used to investigate other maternal infections that impact foetal development.


Asunto(s)
Feto , Placenta , Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika/virología , Virus Zika , Animales , Chlorocebus aethiops , Femenino , Feto/inmunología , Feto/virología , Cobayas , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/inmunología , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Células Vero , Virus Zika/inmunología , Virus Zika/patogenicidad
15.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166244, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34411716

RESUMEN

The placenta provides a significant physical and physiological barrier to prevent fetal infection during pregnancy. Nevertheless, it is at times breached by pathogens and leads to vertical transmission of infection from mother to fetus. This review will focus specifically on the Zika flavivirus, the HIV retrovirus and the emerging SARS-CoV2 coronavirus, which have affected pregnant women and their offspring in recent epidemics. In particular, we will address how viral infections affect the immune response at the maternal-fetal interface and how the placental barrier is physically breached and discuss the consequences of infection on various aspects of placental function to support fetal growth and development. Improved understanding of how the placenta responds to viral infections will lay the foundation for developing therapeutics to these and emergent viruses, to minimise the harms of infection to the offspring.


Asunto(s)
Placenta/virología , Complicaciones Infecciosas del Embarazo/virología , Virosis/fisiopatología , COVID-19/metabolismo , Femenino , Feto/virología , Infecciones por VIH/metabolismo , VIH-1/patogenicidad , Humanos , Transmisión Vertical de Enfermedad Infecciosa/estadística & datos numéricos , Placenta/metabolismo , Embarazo , Complicaciones Infecciosas del Embarazo/epidemiología , SARS-CoV-2/patogenicidad , Virus Zika/patogenicidad , Infección por el Virus Zika/metabolismo
16.
J Virol ; 95(21): e0081821, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34379510

RESUMEN

Zika virus (ZIKV) is a flavivirus that causes a constellation of adverse fetal outcomes collectively termed congenital Zika syndrome (CZS). However, not all pregnancies exposed to ZIKV result in an infant with apparent defects. During the 2015 to 2016 American outbreak of ZIKV, CZS rates varied by geographic location. The underlying mechanisms responsible for this heterogeneity in outcomes have not been well defined. Therefore, we sought to characterize and compare the pathogenic potential of multiple Asian-/American-lineage ZIKV strains in an established Ifnar1-/- pregnant mouse model. Here, we show significant differences in the rate of fetal demise following maternal inoculation with ZIKV strains from Puerto Rico, Panama, Mexico, Brazil, and Cambodia. Rates of fetal demise broadly correlated with maternal viremia but were independent of fetus and placenta virus titer, indicating that additional underlying factors contribute to fetal outcome. Our results, in concert with those from other studies, suggest that subtle differences in ZIKV strains may have important phenotypic impacts. With ZIKV now endemic in the Americas, greater emphasis needs to be placed on elucidating and understanding the underlying mechanisms that contribute to fetal outcome. IMPORTANCE Zika virus (ZIKV) transmission has been reported in 87 countries and territories around the globe. ZIKV infection during pregnancy is associated with adverse fetal outcomes, including birth defects, microcephaly, neurological complications, and even spontaneous abortion. Rates of adverse fetal outcomes vary between regions, and not every pregnancy exposed to ZIKV results in birth defects. Not much is known about how or if the infecting ZIKV strain is linked to fetal outcomes. Our research provides evidence of phenotypic heterogeneity between Asian-/American-lineage ZIKV strains and provides insight into the underlying causes of adverse fetal outcomes. Understanding ZIKV strain-dependent pathogenic potential during pregnancy and elucidating underlying causes of diverse clinical sequelae observed during human infections is critical to understanding ZIKV on a global scale.


Asunto(s)
Feto/patología , Complicaciones Infecciosas del Embarazo/virología , Receptor de Interferón alfa y beta/genética , Infección por el Virus Zika/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Feto/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/virología , Embarazo , Complicaciones Infecciosas del Embarazo/inmunología , Infección por el Virus Zika/congénito
17.
Ann Clin Lab Sci ; 51(4): 570-572, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34452898

RESUMEN

COVID-19 has affected patients of all ages and demographics, not excluding pregnant women. The effects of COVID-19 on pregnant women are still largely unknown. Several adverse perinatal outcomes have been reported in COVID-19-positive pregnant women, including pre-eclampsia, miscarriage, pre-term labor, and stillbirth. Histopathological examination of COVID-19 placentas can contribute significant data regarding maternal and fetal health and can elucidate more findings in this novel disease. A 23-year-old female with morbid obesity and scant antenatal care presented to the emergency department complaining of shortness of breath and fever; she was found to be positive for COVID-19. Grossly, her placenta showed no abnormalities. Histological examination of her placenta showed chronic lymphoplasmacytic deciduitis, villous fibrosis, loss of capillarization, extravasation of erythrocytes, chorangiosis, and thrombosis of upstream stem vessels, including large fetal vessels on the chorionic plate. These changes were deemed to be consistent with fetal thrombotic vasculopathy (FTV). In conclusion, this case of FTV in the placenta of a patient with COVID-19 is a significant finding, as it can be critical to clinicians in the management of prenatal care for expecting mothers during this pandemic.This case was presented at the annual meeting of the Association of Clinical Scientists (ACS) on May 13, 2021.


Asunto(s)
COVID-19/complicaciones , Enfermedades Fetales/patología , Feto/patología , Enfermedades Placentarias/patología , Placenta/patología , SARS-CoV-2/aislamiento & purificación , Trombosis/patología , Adulto , COVID-19/transmisión , COVID-19/virología , Femenino , Enfermedades Fetales/virología , Feto/virología , Humanos , Placenta/virología , Enfermedades Placentarias/virología , Embarazo , Pronóstico , Trombosis/virología , Adulto Joven
18.
J Med Virol ; 93(12): 6788-6793, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34260081

RESUMEN

This study aimed to report a case of mild novel coronavirus disease (COVID-19) in a pregnant woman with probable viremia, as reverse transcription-polymerase chain reaction (RT-PCR) testing of endometrial and placental swabs for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was positive. A 26-year-old multigravida at 35 weeks 2 days of gestation, who had extensive thigh and abdominal cellulitis, tested SARS-CoV-2 positive by RT-PCR performed on samples from the endometrium and maternal side of the placenta. However, other samples (amniotic fluid, fetal side of the placenta, umbilical cord, maternal vagina, and neonatal nasopharynx) tested negative for SARS-CoV-2. This is one of the rare reports of probable SARS-CoV-2 viremia with the presence of SARS-CoV-2 in the endometrium and placenta, but not leading to vertical transmission and neonatal infection. Because knowledge about transplacental transmission and results is very limited, we conclude that more RT-PCR tests on placental and cord blood samples are needed in order to safely make definite conclusions.


Asunto(s)
COVID-19/virología , Feto/virología , Placenta/virología , Complicaciones Infecciosas del Embarazo/virología , SARS-CoV-2/genética , Viremia/virología , Adulto , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Embarazo , Mujeres Embarazadas
19.
J Virol ; 95(17): e0070521, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34260264

RESUMEN

The latest outbreak of Zika virus (ZIKV) in the Americas was associated with significant neurologic complications, including microcephaly of newborns. We evaluated mechanisms that regulate ZIKV entry into human fetal astrocytes (HFAs). Astrocytes are key players in maintaining brain homeostasis. We show that the central mediator of canonical Wnt signaling, ß-catenin, regulates Axl, a receptor for ZIKV infection of HFAs, at the transcriptional level. In turn, ZIKV inhibited ß-catenin, potentially as a mechanism to overcome its restriction of ZIKV internalization through regulation of Axl. This was evident with three ZIKV strains tested but not with a laboratory-adapted strain which has a large deletion in its envelope gene. Finally, we show that ß-catenin-mediated Axl-dependent internalization of ZIKV may be of increased importance for brain cells, as it regulated ZIKV infection of astrocytes and human brain microvascular cells but not kidney epithelial (Vero) cells. Collectively, our studies reveal a role for ß-catenin in ZIKV infection and highlight a dynamic interplay between ZIKV and ß-catenin to modulate ZIKV entry into susceptible target cells. IMPORTANCE ZIKV is an emerging pathogen with sporadic outbreaks throughout the world. The most recent outbreak in North America was associated with small brains (microcephaly) in newborns. We studied the mechanism(s) that may regulate ZIKV entry into astrocytes. Astrocytes are a critical resident brain cell population with diverse functions that maintain brain homeostasis, including neurogenesis and neuronal survival. We show that three ZIKV strains (and not a heavily laboratory-adapted strain with a large deletion in its envelope gene) require Axl for internalization. Most importantly, we show that ß-catenin, the central mediator of canonical Wnt signaling, negatively regulates Axl at the transcriptional level to prevent ZIKV internalization into human fetal astrocytes. To overcome this restriction, ZIKV downregulates ß-catenin to facilitate Axl expression. This highlights a dynamic host-virus interaction whereby ZIKV inhibits ß-catenin to promote its internalization into human fetal astrocytes through the induction of Axl.


Asunto(s)
Astrocitos/virología , Encéfalo/virología , Feto/virología , Replicación Viral , Infección por el Virus Zika/prevención & control , Virus Zika/fisiología , beta Catenina/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Chlorocebus aethiops , Feto/metabolismo , Humanos , Riñón/metabolismo , Riñón/virología , Células Vero , Internalización del Virus , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , beta Catenina/genética
20.
J Infect Dis ; 224(Suppl 6): S647-S659, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34293137

RESUMEN

BACKGROUND: Expression of angiotensin-converting enzyme 2 (ACE2) and type II transmembrane serine protease (TMPRSS2), host molecules required for viral entry, may underlie sex differences in vulnerability to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. We investigated whether placental ACE2 and TMPRSS2 expression vary by fetal sex in the presence of maternal SARS-CoV-2 infection. METHODS: Placental ACE2 and TMPRSS2 expression was quantified by quantitative reverse transcription polymerase chain reaction (RT-PCR) and by Western blot in 68 pregnant women (38 SARS-CoV-2 positive, 30 SARS-CoV-2 negative) delivering at Mass General Brigham from April to June 2020. The impact of fetal sex and maternal SARS-CoV-2 exposure on ACE2 and TMPRSS2 was analyzed by 2-way analysis of variance (ANOVA). RESULTS: Maternal SARS-CoV-2 infection impacted placental TMPRSS2 expression in a sexually dimorphic fashion (2-way ANOVA interaction, P = .002). We observed no impact of fetal sex or maternal SARS-CoV-2 status on ACE2. TMPRSS2 expression was significantly correlated with ACE2 expression in males (Spearman ρ = 0.54, P = .02) but not females (ρ = 0.23, P = .34) exposed to maternal SARS-CoV-2. CONCLUSIONS: Sex differences in placental TMPRSS2 but not ACE2 were observed in the setting of maternal SARS-CoV-2 infection, which may have implications for offspring vulnerability to placental infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/diagnóstico , Sangre Fetal/inmunología , Placenta/metabolismo , SARS-CoV-2/inmunología , Serina Endopeptidasas/metabolismo , Factores Sexuales , Adulto , COVID-19/sangre , Femenino , Sangre Fetal/virología , Feto/virología , Expresión Génica , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Masculino , Embarazo , Complicaciones Infecciosas del Embarazo/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA