Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 736
Filtrar
1.
Cells ; 13(17)2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39273061

RESUMEN

Zika virus (ZIKV) is an arbovirus with maternal, sexual, and TORCH-related transmission capabilities. After 2015, Brazil had the highest number of ZIVK-infected pregnant women who lost their babies or delivered them with Congenital ZIKV Syndrome (CZS). ZIKV triggers an immune defense in the placenta. This immune response counts with the participation of interleukins and transcription factors. Additionally, it has the potential involvement of human endogenous retroviruses (HERVS). Interleukins are immune response regulators that aid immune tolerance and support syncytial structure development in the placenta, where syncytin receptors facilitate vital cell-to-cell fusion events. HERVs are remnants of ancient viral infections that integrate into the genome and produce syncytin proteins crucial for placental development. Since ZIKV can infect trophoblast cells, we analyzed the relationship between ZIKV infection, HERV, interleukin, and transcription factor modulations in the placenta. To investigate the impact of ZIKV on trophoblast cells, we examined two cell types (BeWo and HTR8) infected with ZIKV-MR766 (African) and ZIKV-IEC-Paraíba (Asian-Brazilian) using Taqman and RT2 Profiler PCR Array assays. Our results indicate that early ZIKV infection (24-72 h) does not induce differential interleukins, transcription factors, and HERV expression. However, we show that the expression of a few of these host defense genes appears to be linked independently of ZIKV infection. Future studies involving additional trophoblastic cell lineages and extended infection timelines will illuminate the dynamic interplay between ZIKV, HERVs, interleukins, and transcription factors in the placenta.


Asunto(s)
Retrovirus Endógenos , Interleucinas , Factores de Transcripción , Trofoblastos , Infección por el Virus Zika , Virus Zika , Humanos , Trofoblastos/virología , Trofoblastos/metabolismo , Femenino , Infección por el Virus Zika/virología , Infección por el Virus Zika/genética , Retrovirus Endógenos/genética , Embarazo , Interleucinas/genética , Interleucinas/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Placenta/virología , Placenta/metabolismo , Línea Celular
2.
Wiad Lek ; 77(7): 1434-1455, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39241144

RESUMEN

OBJECTIVE: Aim: To make a systematic review and meta-analysis of published data on the study of histological and immunohistochemical features of the placenta in women who had acute coronavirus infection associated with SARS-CoV-2 ("Covid" placentas) during pregnancy. PATIENTS AND METHODS: Materials and Methods: The search for literature data is based on the PRISMA methodology); the MEDLINE database (PubMed®) was searched using Medical Subject Headings terms from January 2020 to July 2023. The project was registered in the Open Sience Frame (Project Identifier: DOI 10.17605/OSF.IO/GDR3S, Registration DOI: https://doi.org/10.17605/OSF.IO/H2KPU). Preference was given to studies in which the description of placentas met the requirements of the Amsterdam Placental Workshop Group Consensus Statement. RESULTS: Results: A total of 31 studies were included; the number of participants whose morphological and histological description of the placentas could be subjected to meta-analysis was 2401, respectively, in the group with a "Covid" history and 1910 - conditionally healthy pregnant women. Pathological changes in the placental complex were not detected in 42±19.62% of pregnant women with a history of Covid. Immunohistochemical examination of placentas preferably focuses on the detection of SARS-CoV-2 spike protein or ACE2. According to currently available studies, in the placentas of women who have had COVID-19 during pregnancy, there are no pathognomic histological patterns specific to this infection and direct damage to the placenta is rarely observed. Histological patterns in "covid" placentas are isolated, most often a combination of lesions in both the maternal and fetal malperfusion. CONCLUSION: Conclusions: According to currently available studies, in the placentas of women who have had COVID-19 during pregnancy, there are no pathognomic histological patterns specific to this infection and direct damage to the placenta is rarely observed. The probability of infection of the intrauterine fetus by the transplacental hematogenous route is the lowest compared to other routes, which, in our opinion, is a possible explanation for the high frequency of MVM without subsequent infection of the fetus.


Asunto(s)
COVID-19 , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Humanos , Embarazo , Femenino , COVID-19/patología , Placenta/patología , Placenta/virología , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Inmunohistoquímica , Pandemias , Infecciones por Coronavirus/patología
3.
Virol J ; 21(1): 209, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39227837

RESUMEN

BACKGROUND: Early pregnancy Zika virus (ZIKV) infection is associated with major brain damage in fetuses, leading to microcephaly in 0.6-5.0% of cases, but the underlying mechanisms remain largely unknown. METHODS: To understand the kinetics of ZIKV infection during fetal development in a nonhuman primate model, four cynomolgus macaque fetuses were exposed in utero through echo-guided intramuscular inoculation with 103 PFU of ZIKV at 70-80 days of gestation, 2 controls were mock inoculated. Clinical, immuno-virological and ultrasound imaging follow-ups of the mother/fetus pairs were performed until autopsy after cesarean section 1 or 2 months after exposure (n = 3 per group). RESULTS: ZIKV was transmitted from the fetus to the mother and then replicate in the peripheral blood of the mother from week 1 to 4 postexposure. Infected fetal brains tended to be smaller than those of controls, but not the femur lengths. High level of viral RNA ws found after the first month in brain tissues and placenta. Thereafter, there was partial control of the virus in the fetus, resulting in a decreased number of infected tissue sections and a decreased viral load. Immune cellular and humoral responses were effectively induced. CONCLUSIONS: ZIKV infection during the second trimester of gestation induces short-term brain injury, and although viral genomes persist in tissues, most of the virus is cleared before delivery.


Asunto(s)
Encéfalo , Modelos Animales de Enfermedad , Feto , Complicaciones Infecciosas del Embarazo , Carga Viral , Infección por el Virus Zika , Virus Zika , Animales , Femenino , Embarazo , Infección por el Virus Zika/virología , Feto/virología , Complicaciones Infecciosas del Embarazo/virología , Encéfalo/virología , Macaca fascicularis/virología , ARN Viral , Placenta/virología , Transmisión Vertical de Enfermedad Infecciosa
4.
Schweiz Arch Tierheilkd ; 166(9): 460-464, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39225507

RESUMEN

INTRODUCTION: The novel Porcine circovirus 3 (PCV-3) has been associated in the past years to different porcine diseases, including reproductive failure. The potential occurrence of PCV-3 in abortions from Swiss pig herds has not been investigated so far. Thus, we conducted a retrospective study on pig aborted cases submitted to our laboratory in the University of Bern during the last 10 years with the main aim of investigating the possible presence of PCV-3 in foetal and/or placental tissue. Twelve out of the 53 studied cases showed mild histopathological changes as previously described in PCV-3 positive cases. However, in none of the cases, PCV-3 genetic material could be detected in the examined formalin-fixed, paraffin-embedded tissues. In only one third of the cases, a cause for the abortion was found, which is similar to other studies. Our survey suggests that PCV-3 was not involved in the porcine abortion cases submitted over the last decade at our institution in Switzerland.


INTRODUCTION: Le nouveau Circovirus porcin 3 (PCV-3) a été associé ces dernières années à différentes maladies porcines, y compris des troubles de la reproduction. La présence potentielle du PCV-3 dans les avortements de porcs en Suisse n'a pas été étudiée jusqu'à présent. Nous avons donc mené une étude rétrospective sur les cas d'avortements de porcs soumis à notre laboratoire de l'Université de Berne au cours des 10 dernières années, dans le but principal d'étudier la présence éventuelle du PCV-3 dans les tissus fœtaux et/ou placentaires. Douze des 53 cas étudiés présentaient des changements histopathologiques légers, tels que décrits précédemment dans les cas positifs au PCV-3. Cependant, dans aucun des cas, le matériel génétique du PCV-3 n'a pu être détecté dans les tissus examinés fixés au formol et inclus en paraffine. Dans un tiers des cas seulement, une cause d'avortement a été trouvée, ce qui est similaire à d'autres études. Notre étude suggère que le PCV-3 n'a pas été impliqué dans les cas d'avortements porcins soumis au cours de la dernière décennie dans notre institution en Suisse.


Asunto(s)
Aborto Veterinario , Infecciones por Circoviridae , Circovirus , Enfermedades de los Porcinos , Animales , Femenino , Embarazo , Aborto Veterinario/virología , Infecciones por Circoviridae/veterinaria , Infecciones por Circoviridae/virología , Circovirus/aislamiento & purificación , Circovirus/genética , Formaldehído , Adhesión en Parafina/veterinaria , Placenta/virología , Placenta/patología , Estudios Retrospectivos , Porcinos , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/patología , Suiza/epidemiología
5.
Am J Reprod Immunol ; 92(2): e13911, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39113636

RESUMEN

BACKGROUND: The severe acute respiratory syndrome coronavirus (SARS-CoV-2) outbreak in 2019 has necessitated investigating its potential adverse effects on pregnancy outcomes and fetal development. OBJECTIVE: This study aimed to review the evidence on the impact of SARS-CoV-2 infection during pregnancy on fetal outcomes. METHOD OF STUDY: Literatures since the outbreak of COVID-19 from PubMed and Web of Science were summarized in this narrative review, to show the effects of maternal SARS-CoV-2 infection during pregnancy on fetal development. RESULTS: SARS-CoV-2 infection during pregnancy can be transmitted vertically through the placenta, both in utero and perinatally, affecting the maternal-fetal immune interface and placental function. Viral infections during pregnancy have been linked to central nervous system development impairments and disorders such as autism. Changes in the structure and function of the respiratory, immune, and visceral systems have also been reported. SARS-CoV-2 infection during pregnancy has been linked with increased risks of stillbirth and preterm birth. However, the mechanisms involved remain unclear and may include cytokine storms, macrophage mediation, genetic mutations, methylation, and other epigenetic changes. Exploring the protective effects of antiviral treatment and other interventions in animal and clinical studies may help improve outcomes. CONCLUSION: SARS-CoV-2 infection during pregnancy activates the maternal-fetal immune interface through vertical transmission, and has short- and long-term effects on fetal development, including the central nervous system. Future long-term studies may help provide evidence that can inform interventions to reduce the risk of adverse outcomes.


Asunto(s)
COVID-19 , Desarrollo Fetal , Transmisión Vertical de Enfermedad Infecciosa , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Humanos , Embarazo , COVID-19/inmunología , COVID-19/transmisión , Femenino , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , SARS-CoV-2/inmunología , Desarrollo Fetal/inmunología , Nacimiento Prematuro/inmunología , Placenta/virología , Placenta/inmunología , Resultado del Embarazo
6.
Can Vet J ; 65(8): 813-816, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39091477

RESUMEN

Objective: Our objective was to determine whether equine herpesviruses 1 (EHV-1) viral nucleic acids could be detected immediately after foaling from nasal and vaginal swabs, whole blood, and placental tissue of healthy mares. Animals procedure and results: Nasal and vaginal swabs, EDTA blood, and placental tissue (296 samples) were collected from 74 clinically healthy postpartum broodmares within 24 h after giving birth to live, clinically healthy foals. All samples were tested (PCR) for nucleic acids of neuropathogenic and non-neuropathogenic strains of EHV-1, and all were negative. Conclusion and clinical relevance: As EHV-1 was not detected in the immediate postpartum period in healthy mares with uncomplicated foaling, we inferred that EHV-1-positive samples from aborting mares and/or EHV-1 detection in fetal membranes indicate EHV-1-associated abortion.


Tests moléculaires pour l'herpèsvirus équin 1 (EHV-1) chez des juments poulinières post-partum en bonne santé. Objectif: Notre objectif était de déterminer si les acides nucléiques viraux de l'herpèsvirus équin 1 (EHV-1) pouvaient être détectés immédiatement après la mise bas à partir de prélèvements nasaux et vaginaux, de sang total et de tissus placentaires de juments saines. Animaux procédure et résultats: Des écouvillons nasaux et vaginaux, du sang EDTA et du tissu placentaire (296 échantillons) ont été prélevés sur 74 juments poulinières post-partum cliniquement saines dans les 24 heures suivant la naissance de poulains vivants et cliniquement sains. Tous les échantillons ont été testés (PCR) pour les acides nucléiques des souches neuropathogènes et non-neuropathogènes de l'EHV-1, et tous se sont révélés négatifs. Conclusion et pertinence clinique: Comme l'EHV-1 n'a pas été détecté dans la période post-partum immédiate chez des juments en bonne santé avec un poulinage sans complication, nous avons déduit que les échantillons positifs pour l'EHV-1 provenant de juments qui ont avorté et/ou la détection de l'EHV-1 dans les membranes foetales indiquent un avortement associé à l'EHV-1.(Traduit par Dr Serge Messier).


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Équido 1 , Enfermedades de los Caballos , Periodo Posparto , Animales , Caballos , Herpesvirus Équido 1/aislamiento & purificación , Femenino , Enfermedades de los Caballos/virología , Enfermedades de los Caballos/diagnóstico , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/diagnóstico , Embarazo , Placenta/virología , Vagina/virología , Aborto Veterinario/virología , ADN Viral/análisis , ADN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa/veterinaria
7.
Curr Opin Infect Dis ; 37(5): 402-406, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39105669

RESUMEN

PURPOSE OF REVIEW: To understand the characteristics and determinants of transplacental antibody transfer against SARS-CoV-2 and to compare the differences between SARS-CoV-2 infection and vaccination. RECENT FINDINGS: The need for information during the COVID-19 pandemic and the exclusion of pregnant women from randomized clinical trials have led to a vast amount of clinical data primarily based on observational studies with diverse design and sample analyses that yield variable results. This review aims to critically and comprehensively integrate the relevant knowledge related to transplacental transfer of antibodies against SARS-CoV-2, emphasizing the differences between infection and vaccination. SUMMARY: Passive immunization is key to conferring protection to the infant during their first months of life. Understanding the mechanisms of transplacental antibody transfer during SARS-CoV-2 infection and vaccination, and their associated protection will allow optimizing the implementation of well tolerated and effective preventive strategies for both pregnant women and infants.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Vacunación , Humanos , Embarazo , Femenino , COVID-19/prevención & control , COVID-19/inmunología , SARS-CoV-2/inmunología , Complicaciones Infecciosas del Embarazo/prevención & control , Complicaciones Infecciosas del Embarazo/inmunología , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/administración & dosificación , Placenta/virología , Placenta/inmunología , Vacunación/métodos , Inmunidad Materno-Adquirida , Anticuerpos Antivirales/inmunología , Inmunización Pasiva/métodos
8.
Int J Mol Sci ; 25(16)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39201511

RESUMEN

It has been shown that vertical transmission of the SARS-CoV-2 strain is relatively rare, and there is still limited information on the specific impact of maternal SARS-CoV-2 infection on vertical transmission. The current study focuses on a transcriptomics analysis aimed at examining differences in gene expression between placentas from mother-newborn pairs affected by COVID-19 and those from unaffected controls. Additionally, it investigates the in situ expression of molecules involved in placental inflammation. The Papa Giovanni XXIII Hospital in Bergamo, Italy, has recorded three instances of intrauterine transmission of SARS-CoV-2. The first two cases occurred early in the pandemic and involved pregnant women in their third trimester who were diagnosed with SARS-CoV-2. The third case involved an asymptomatic woman in her second trimester with a twin pregnancy, who unfortunately delivered two stillborn fetuses due to the premature rupture of membranes. Transcriptomic analysis revealed significant differences in gene expression between the placentae of COVID-19-affected mother/newborn pairs and two matched controls. The infected and control placentae were matched for gestational age. According to the Benjamani-Hochberg method, 305 genes met the criterion of an adjusted p-value of less than 0.05, and 219 genes met the criterion of less than 0.01. Up-regulated genes involved in cell signaling (e.g., CCL20, C3, MARCO) and immune response (e.g., LILRA3, CXCL10, CD48, CD86, IL1RN, IL-18R1) suggest their potential role in the inflammatory response to SARS-CoV-2. RNAscope® technology, coupled with image analysis, was utilized to quantify the surface area covered by SARS-CoV-2, ACE2, IL-1ß, IL-6, IL-8, IL-10, and TNF-α on both the maternal and fetal sides of the placenta. A non-statistically significant gradient for SARS-CoV-2 was observed, with a higher surface coverage on the fetal side (2.42 ± 3.71%) compared to the maternal side (0.74 ± 1.19%) of the placenta. Although not statistically significant, the surface area covered by ACE2 mRNA was higher on the maternal side (0.02 ± 0.04%) compared to the fetal side (0.01 ± 0.01%) of the placenta. IL-6 and IL-8 were more prevalent on the fetal side (0.03 ± 0.04% and 0.06 ± 0.08%, respectively) compared to the maternal side (0.02 ± 0.01% and 0.02 ± 0.02%, respectively). The mean surface areas of IL-1ß and IL-10 were found to be equal on both the fetal (0.04 ± 0.04% and 0.01 ± 0.01%, respectively) and maternal sides of the placenta (0.04 ± 0.05% and 0.01 ± 0.01%, respectively). The mean surface area of TNF-α was found to be equal on both the fetal and maternal sides of the placenta (0.02 ± 0.02% and 0.02 ± 0.02%, respectively). On the maternal side, ACE-2 and all examined interleukins, but not TNF-α, exhibited an inverse mRNA amount compared to SARS-CoV-2. On the fetal side, ACE-2, IL-6 and IL-8 were inversely correlated with SARS-CoV-2 (r = -0.3, r = -0.1 and r = -0.4, respectively), while IL-1ß and IL-10 showed positive correlations (r = 0.9, p = 0.005 and r = 0.5, respectively). TNF-α exhibited a positive correlation with SARS-CoV-2 on both maternal (r = 0.4) and fetal sides (r = 0.9) of the placenta. Further research is needed to evaluate the correlation between cell signaling and immune response genes in the placenta and the vertical transmission of SARS-CoV-2. Nonetheless, the current study extends our comprehension of the molecular and immunological factors involved in SARS-CoV-2 placental infection underlying maternal-fetal transmission.


Asunto(s)
COVID-19 , Transmisión Vertical de Enfermedad Infecciosa , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Adulto , Femenino , Humanos , Recién Nacido , Embarazo , COVID-19/inmunología , COVID-19/transmisión , COVID-19/virología , Citocinas/metabolismo , Citocinas/genética , Perfilación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/virología , Placenta/inmunología , Placenta/metabolismo , Placenta/virología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , SARS-CoV-2/inmunología , Transcriptoma
9.
J Virol ; 98(8): e0098324, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39016561

RESUMEN

Rift Valley fever virus (RVFV) infection causes abortions in ruminant livestock and is associated with an increased likelihood of miscarriages in women. Using sheep and human placenta explant cultures, we sought to identify tissues at the maternal-fetal interface targeted by RVFV. Sheep villi and fetal membranes were highly permissive to RVFV infection resulting in markedly higher virus titers than human cultures. Sheep cultures were most permissive to wild-type RVFV and ΔNSm infection, while live-attenuated RVFV vaccines (LAVs; MP-12, ΔNSs, and ΔNSs/ΔNSm) exhibited reduced replication. The human fetal membrane restricted wild-type and LAV replication, and when infection occurred, it was prominent on the maternal-facing side. Type I and type III interferons were induced in human villi exposed to LAVs lacking the NSs protein. This study supports the use of sheep and human placenta explants to understand vertical transmission of RVFV in mammals and whether LAVs are attenuated at the maternal-fetal interface.IMPORTANCEA direct comparison of replication of Rift Valley fever virus (RVFV) in sheep and human placental explants reveals comparative efficiencies and permissivity to infection and replication. Vaccine strains of RVFV demonstrated reduced infection and replication capacity in the mammalian placenta. This study represents the first direct cross-host comparison of the vertical transmission capacity of this high-priority emerging mosquito-transmitted virus.


Asunto(s)
Transmisión Vertical de Enfermedad Infecciosa , Placenta , Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift , Vacunas Atenuadas , Vacunas Virales , Replicación Viral , Virus de la Fiebre del Valle del Rift/fisiología , Virus de la Fiebre del Valle del Rift/inmunología , Animales , Femenino , Embarazo , Ovinos , Placenta/virología , Humanos , Fiebre del Valle del Rift/virología , Fiebre del Valle del Rift/transmisión , Vacunas Virales/inmunología , Enfermedades de las Ovejas/virología
10.
PLoS Pathog ; 20(7): e1012408, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39038037

RESUMEN

c-FLIP functions as a dual regulator of apoptosis and inflammation, yet its implications in Zika virus (ZIKV) infection remain partially understood, especially in the context of ZIKV-induced congenital Zika syndrome (CZS) where both apoptosis and inflammation play pivotal roles. Our findings demonstrate that c-FLIP promotes ZIKV infection in placental cells and myeloid-derived macrophages, involving inflammation and caspase-8/3-mediated apoptosis. Moreover, our observations reveal that c-FLIP augments ZIKV infection in multiple tissues, including blood cell, spleen, uterus, testis, and the brain of mice. Notably, the partial deficiency of c-FLIP provides protection to embryos against ZIKV-induced CZS, accompanied by a reduction in caspase-3-mediated apoptosis. Additionally, we have found a distinctive parental effect of c-FLIP influencing ZIKV replication in fetal heads. In summary, our study reveals the critical role of c-FLIP as a positive regulator in caspase-8/3-mediated apoptosis during ZIKV infection, significantly contributing to the development of CZS.


Asunto(s)
Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Caspasa 3 , Caspasa 8 , Infección por el Virus Zika , Virus Zika , Infección por el Virus Zika/virología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/patología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Animales , Ratones , Caspasa 8/metabolismo , Femenino , Humanos , Caspasa 3/metabolismo , Embarazo , Placenta/virología , Placenta/metabolismo , Placenta/patología , Ratones Endogámicos C57BL , Replicación Viral , Ratones Noqueados
11.
J Exp Med ; 221(9)2024 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-39042188

RESUMEN

The contribution of placental immune responses to congenital Zika virus (ZIKV) syndrome remains poorly understood. Here, we leveraged a mouse model of ZIKV infection to identify mechanisms of innate immune restriction exclusively in the fetal compartment of the placenta. ZIKV principally infected mononuclear trophoblasts in the junctional zone, which was limited by mitochondrial antiviral-signaling protein (MAVS) and type I interferon (IFN) signaling mechanisms. Single nuclear RNA sequencing revealed MAVS-dependent expression of IFN-stimulated genes (ISGs) in spongiotrophoblasts but not in other placental cells that use alternate pathways to induce ISGs. ZIKV infection of Ifnar1-/- or Mavs-/- placentas was associated with greater infection of the adjacent immunocompetent decidua, and heterozygous Mavs+/- or Ifnar1+/- dams carrying immunodeficient fetuses sustained greater maternal viremia and tissue infection than dams carrying wild-type fetuses. Thus, MAVS-IFN signaling in the fetus restricts ZIKV infection in junctional zone trophoblasts, which modulates dissemination and outcome for both the fetus and the pregnant mother.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Decidua , Feto , Interferón Tipo I , Placenta , Receptor de Interferón alfa y beta , Transducción de Señal , Trofoblastos , Infección por el Virus Zika , Virus Zika , Femenino , Animales , Embarazo , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Placenta/inmunología , Placenta/virología , Placenta/metabolismo , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología , Virus Zika/inmunología , Virus Zika/fisiología , Ratones , Decidua/inmunología , Decidua/virología , Decidua/metabolismo , Feto/inmunología , Feto/virología , Trofoblastos/inmunología , Trofoblastos/virología , Trofoblastos/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Inmunidad Innata , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/virología , Modelos Animales de Enfermedad
12.
J Med Virol ; 96(7): e29819, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39030992

RESUMEN

Pregnant women represent a high-risk population for Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) infection. The presence of SARS-CoV-2 has been reported in placenta from infected pregnant women, but whether the virus influences placenta immune response remains unclear. We investigated the properties of maternal-fetal interface macrophages (MFMs) in a cohort of unvaccinated women who contracted coronavirus disease 2019 (COVID-19) during their pregnancy. We reported an infiltration of CD163+ macrophages in placenta from COVID-19 women 19 whereas lymphoid compartment was not affected. Isolated MFMs exhibited nonpolarized activated signature (NOS2, IDO1, IFNG, TNF, TGFB) mainly in women infected during the second trimester of pregnancy. COVID-19 during pregnancy primed MFM to produce type I and III interferon response to SARS-CoV-2 (Wuhan and δ strains), that were unable to elicit this in MFMs from healthy pregnant women. COVID-19 also primed SARS-CoV-2 internalization by MFM in an angiotensin-converting enzyme 2-dependent manner. Activation and recall responses of MFMs were influenced by fetal sex. Collectively, these findings support a role for MFMs in the local immune response to SARS-CoV-2 infection, provide a basis for protective placental immunity in COVID-19, and highlight the interest of vaccination in pregnant women.


Asunto(s)
COVID-19 , Macrófagos , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Humanos , Femenino , Embarazo , COVID-19/inmunología , COVID-19/virología , Placenta/inmunología , Placenta/virología , Macrófagos/inmunología , Macrófagos/virología , Complicaciones Infecciosas del Embarazo/virología , Complicaciones Infecciosas del Embarazo/inmunología , SARS-CoV-2/inmunología , Adulto , Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Internalización del Virus
13.
FASEB J ; 38(13): e23799, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38979938

RESUMEN

Maternal Zika virus (ZIKV) infection during pregnancy has been associated with severe intrauterine growth restriction (IUGR), placental damage, metabolism disturbances, and newborn neurological abnormalities. Here, we investigated the impact of maternal ZIKV infection on placental nutrient transporters and nutrient-sensitive pathways. Immunocompetent (C57BL/6) mice were injected with Low (103 PFU-ZIKVPE243) or High (5 × 107 PFU-ZIKVPE243) ZIKV titers at gestational day (GD) 12.5, and tissue was collected at GD18.5 (term). Fetal-placental growth was impaired in male fetuses, which exhibited higher placental expression of the ZIKV infective marker, eukaryotic translation initiation factor 2 (eIF2α), but lower levels of phospho-eIF2α. There were no differences in fetal-placental growth in female fetuses, which exhibited no significant alterations in placental ZIKV infective markers. Furthermore, ZIKV promoted increased expression of glucose transporter type 1 (Slc2a1/Glut1) and decreased levels of glucose-6-phosphate in female placentae, with no differences in amino acid transport potential. In contrast, ZIKV did not impact glucose transporters in male placentae but downregulated sodium-coupled neutral amino acid 2 (Snat2) transporter expression. We also observed sex-dependent differences in the hexosamine biosynthesis pathway (HBP) and O-GlcNAcylation in ZIKV-infected pregnancies, showing that ZIKV can disturb placental nutrient sensing. Our findings highlight molecular alterations in the placenta caused by maternal ZIKV infection, shedding light on nutrient transport, sensing, and availability. Our results also suggest that female and male placentae employ distinct coping mechanisms in response to ZIKV-induced metabolic changes, providing insights into therapeutic approaches for congenital Zika syndrome.


Asunto(s)
Desarrollo Fetal , Ratones Endogámicos C57BL , Placenta , Transducción de Señal , Infección por el Virus Zika , Virus Zika , Animales , Femenino , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología , Embarazo , Ratones , Placenta/metabolismo , Placenta/virología , Masculino , Desarrollo Fetal/fisiología , Complicaciones Infecciosas del Embarazo/virología , Complicaciones Infecciosas del Embarazo/metabolismo , Nutrientes/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo
14.
Viruses ; 16(6)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38932210

RESUMEN

Human cytomegalovirus (CMV) infection is the leading non-genetic cause of congenital malformation in developed countries, causing significant fetal injury, and in some cases fetal death. The pathogenetic mechanisms through which this host-specific virus infects then damages both the placenta and the fetal brain are currently ill-defined. We investigated the CMV modulation of key signaling pathway proteins for these organs including dual-specificity tyrosine phosphorylation-regulated kinases (DYRK) and Sonic Hedgehog (SHH) pathway proteins using human first trimester placental trophoblast (TEV-1) cells, primary human astrocyte (NHA) brain cells, and CMV-infected human placental tissue. Immunofluorescence demonstrated the accumulation and re-localization of SHH proteins in CMV-infected TEV-1 cells with Gli2, Ulk3, and Shh re-localizing to the CMV cytoplasmic virion assembly complex (VAC). In CMV-infected NHA cells, DYRK1A re-localized to the VAC and DYRK1B re-localized to the CMV nuclear replication compartments, and the SHH proteins re-localized with a similar pattern as was observed in TEV-1 cells. Western blot analysis in CMV-infected TEV-1 cells showed the upregulated expression of Rb, Ulk3, and Shh, but not Gli2. In CMV-infected NHA cells, there was an upregulation of DYRK1A, DYRK1B, Gli2, Rb, Ulk3, and Shh. These in vitro monoculture findings are consistent with patterns of protein upregulation and re-localization observed in naturally infected placental tissue and CMV-infected ex vivo placental explant histocultures. This study reveals CMV-induced changes in proteins critical for fetal development, and identifies new potential targets for CMV therapeutic development.


Asunto(s)
Astrocitos , Infecciones por Citomegalovirus , Citomegalovirus , Proteínas Hedgehog , Placenta , Proteínas Tirosina Quinasas , Transducción de Señal , Humanos , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Citomegalovirus/fisiología , Embarazo , Placenta/virología , Placenta/metabolismo , Astrocitos/virología , Astrocitos/metabolismo , Femenino , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/genética , Infecciones por Citomegalovirus/virología , Infecciones por Citomegalovirus/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Fosforilación , Trofoblastos/virología , Trofoblastos/metabolismo , Quinasas DyrK , Línea Celular , Células Cultivadas
15.
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
16.
J Neuroinflammation ; 21(1): 163, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918792

RESUMEN

BACKGROUND: The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. METHODS AND RESULTS: We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls. CONCLUSION: These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.


Asunto(s)
COVID-19 , Macrófagos , Microglía , Placenta , Complicaciones Infecciosas del Embarazo , SARS-CoV-2 , Femenino , Embarazo , Microglía/virología , Humanos , Placenta/virología , COVID-19/inmunología , Macrófagos/virología , Complicaciones Infecciosas del Embarazo/virología , Complicaciones Infecciosas del Embarazo/patología , SARS-CoV-2/patogenicidad , Feto , Adulto , Encéfalo/virología , Encéfalo/patología , Ratones , Animales
17.
Biochem Biophys Res Commun ; 726: 150281, 2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-38909532

RESUMEN

Cell-fusion mediated generation of multinucleated syncytia represent critical feature during viral infection and in development. Efficiency of syncytia formation is usually illustrated as fusion efficiency under given condition by quantifying total number of nuclei in syncytia normalized to total number of nuclei (both within syncytia and unfused cell nuclei) in unit field of view. However heterogeneity in multinucleated syncytia sizes poses challenge in quantification of cell-fusion multinucleation under diverse conditions. Taking in-vitro SARS-CoV-2 spike-protein variants mediated virus-cell fusion model and placenta trophoblast syncytialization as cell-cell fusion model; herein we emphasize wide application of simple unbiased detailed measure of virus-cell and cell-cell multinucleation using experiential cumulative distribution function (CDF) and fusion number events (FNE) approaches illustrating comprehensive metrics for syncytia interpretation.


Asunto(s)
Fusión Celular , Células Gigantes , SARS-CoV-2 , Trofoblastos , Humanos , Células Gigantes/virología , Células Gigantes/citología , SARS-CoV-2/fisiología , Trofoblastos/virología , Trofoblastos/citología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Femenino , COVID-19/virología , Embarazo , Internalización del Virus , Placenta/virología , Placenta/citología
18.
Sci Rep ; 14(1): 14605, 2024 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-38918446

RESUMEN

A previous study suggested that fetal inheritance of chromosomally integrated human herpesvirus 6 (ici-HHV6) is associated with the hypertensive pregnancy disorder preeclampsia (PE). We aimed to study this question utilizing cord plasma samples (n = 1276) of the Finnish Genetics of Preeclampsia Consortium (FINNPEC) cohort: 539 from a pregnancy with PE and 737 without. We studied these samples and 30 placentas from PE pregnancies by a multiplex qPCR for the DNAs of all nine human herpesviruses. To assess the population prevalence of iciHHV-6, we studied whole-genome sequencing data from blood-derived DNA of 3421 biobank subjects. Any herpes viral DNA was detected in only two (0.37%) PE and one (0.14%) control sample (OR 2.74, 95% CI 0.25-30.4). One PE sample contained iciHHV-6B and another HHV-7 DNA. The control's DNA was of iciHHV-6B; the fetus having growth restriction and preterm birth without PE diagnosis. Placentas showed no herpesviruses. In the biobank data, 3 of 3421 subjects (0.08%) had low level HHV-6B but no iciHHV-6. While iciHHV-6 proved extremely rare, both fetuses with iciHHV-6B were growth-restricted, preterm, and from a pregnancy with maternal hypertension. Our findings suggest that human herpesviruses are not a significant cause of PE, whereas iciHHV-6 may pose some fetal risk.


Asunto(s)
Herpesvirus Humano 6 , Preeclampsia , Humanos , Femenino , Embarazo , Preeclampsia/virología , Preeclampsia/epidemiología , Adulto , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/aislamiento & purificación , Estudios de Cohortes , Sangre Fetal/virología , Finlandia/epidemiología , ADN Viral/genética , ADN Viral/sangre , Placenta/virología , Herpesviridae/genética
19.
Placenta ; 152: 23-30, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38768555

RESUMEN

INTRODUCTION: Knowledge on prevalence and association of human papillomavirus (HPV) in third trimester placentae and adverse pregnancy outcomes is limited. We investigated the prevalence of placental HPV at delivery, explored urine HPV characteristics associated with placental HPV and whether placental HPV increased the risk adverse pregnancy outcomes. METHODS: Pregnant women were enrolled in the Scandinavian PreventADALL mother-child cohort study at midgestation. Human papillomavirus genotyping was performed on placental biopsies collected at delivery (n = 587) and first-void urine at midgestation and delivery (n = 556). Maternal characteristics were collected by questionnaires at gestational week 18 and 34. Adverse pregnancy outcomes were registered from chart data including hypertensive disorders of pregnancy, gestational diabetes mellitus and newborns small for gestational age. Uni- and multivariable regression models were used to investigate associations. RESULTS: Placental HPV was detected in 18/587 (3 %). Twenty-eight genotypes were identified among the 214/556 (38 %) with midgestational urine HPV. Seventeen of the 18 women with placental HPV were midgestational HPV positive with 89 % genotype concordance. Midgestational high-risk-(HR)-HPV and high viral loads of Any- or HR-HPV were associated with placental HPV. Persisting HPV infection from midgestation to delivery was not associated with placental HPV. Adverse pregnancy outcomes were seen in 2/556 (0.4 %) of women with placental HPV. DISCUSSION: In this general cohort of pregnant women, the prevalence of placental HPV was 3 %, and midgestational urinary HPV 38 %. High HPV viral load increased the risk for placental HPV infections. We observed no increased risk for adverse pregnancy outcomes in women with placental HPV.


Asunto(s)
Infecciones por Papillomavirus , Placenta , Complicaciones Infecciosas del Embarazo , Resultado del Embarazo , Humanos , Femenino , Embarazo , Infecciones por Papillomavirus/epidemiología , Infecciones por Papillomavirus/virología , Adulto , Placenta/virología , Complicaciones Infecciosas del Embarazo/epidemiología , Complicaciones Infecciosas del Embarazo/virología , Papillomaviridae/genética , Estudios de Cohortes , Tercer Trimestre del Embarazo , Adulto Joven
20.
Placenta ; 152: 39-52, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38788480

RESUMEN

INTRODUCTION: Several factors influence transmission of 2019-nCoV from mother to fetus during pregnancy, thus the dynamics of vertical transmission is unclear. The role of cellular protective factors, namely a 90 KDa glycoprotein, Early pregnancy-associated protein (Epap-1), expressed by placental endothelial cells in women during early pregnancy would provide an insight into role of placental factors in virus transmission. Since viral spike protein binding to the ACE2 receptors of the host cells promotes virus invasion in placental tissue, an analysis of effects of Epap-1 on the Spike-ACE2 protein binding was studied. METHODS: Epap-1 was isolated from MTP placental tissue. Molecular interaction of Epap-1 and variants of the spike was analyzed in silco. The interaction of Epap-1 with Spike and RBD were analyzed using ELISA and immunofluorescence studies. RESULTS: The results in silico showed an interaction of Epap-1 with S-protein at RBD region involving K417, Y449, Y453, Y456, Y473, Q474, F486, Q498, N501 residues of spike with Y61, F287, I302, N303, N305, S334, N465, G467, N468 residues of Epap-1 leading to interference of S-protein and ACE2 interaction [1]. Further, the interaction is conserved among the variants. The studies in vitro confirm that Epap-1 affects S protein-ACE2 and RBD- ACE2 binding, thus suggesting that during early pregnancy, SARS CoV-2 infection may be protected by Epap-1 protein present in placental tissue. The results were further confirmed by pseudovirus expressing Spike and RBD in an infection assay. DISCUSSION: Epap-1 interferes with Spike and RBD interaction with ACE2, suggesting a possible mechanism of the antiviral environment during pregnancy.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Transmisión Vertical de Enfermedad Infecciosa , Placenta , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Femenino , Humanos , Embarazo , Enzima Convertidora de Angiotensina 2/metabolismo , Betacoronavirus/metabolismo , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , COVID-19/transmisión , COVID-19/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Placenta/metabolismo , Placenta/virología , Neumonía Viral/metabolismo , Neumonía Viral/transmisión , Neumonía Viral/virología , Complicaciones Infecciosas del Embarazo/metabolismo , Complicaciones Infecciosas del Embarazo/virología , Proteínas Gestacionales/metabolismo , Unión Proteica , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA