Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Front Immunol ; 13: 1031248, 2022.
Article in English | MEDLINE | ID: covidwho-2198876

ABSTRACT

Background: Limited data are available regarding the differences between immunological, biochemical, and cellular contents of human colostrum following maternal infection during pregnancy with coronavirus 2 disease (COVID-19). Objective: To investigate whether maternal COVID-19 infection may affect immunological, biochemical, and cellular contents of human colostrum. Methods: Using a case-control study design, we collected colostrum from 14 lactating women with a previous diagnosis of COVID-19 during pregnancy and 12 without a clear diagnosis during September 2020 to May 2021. Colostrum samples were analysed for some enzymes and non-enzymatic oxidative stress markers (SOD, CAT, GPx, MDA, GSH, GSSG, H2O2, MPO) and for IL-1ß, IL-6, tumour necrosis factor (TNF)-α, protein induced by interferon gamma (IP)-10, IL-8, IFN-λ1, IL12p70, IFN-α2, IFN-λ2/3, granulocyte macrophage colony stimulating factor (GM-CSF), IFN-ß, IL-10 and IFN-γ, along with IgA and IgG for the SARS-CoV-2 S protein. We perform immunophenotyping to assess the frequency of different cell types in the colostrum. Results: Colostrum from the COVID-19 symptomatic group in pregnancy contained reduced levels of H2O2, IFN-α2, and GM-CSF. This group had higher levels of GSH, and both NK cell subtypes CD3-CD56brightCD16-CD27+IFN-γ+ and CD3-CD56dimCD16+CD27- were also increased. Conclusion: The present results reinforce the protective role of colostrum even in the case of mild SARS-Cov-2 infection, in addition to demonstrating how adaptive the composition of colostrum is after infections. It also supports the recommendation to encourage lactating women to continue breastfeeding after COVID-19 illness.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Pregnancy , Female , Humans , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Colostrum/metabolism , COVID-19/metabolism , Case-Control Studies , Hydrogen Peroxide/metabolism , Lactation , SARS-CoV-2 , Interferon-gamma/metabolism , Pregnancy Complications, Infectious/metabolism
2.
Placenta ; 132: 38-43, 2023 02.
Article in English | MEDLINE | ID: covidwho-2165764

ABSTRACT

INTRODUCTION: Recent studies reported a differential expression of both ACE2 and CD147 in pregnant women associated to SARS-CoV-2 placental infection. The aim of this study is to further investigate the placental SARS-CoV-2 infection and the potential effect on protein expression (ACE2, CD147, HLA-G and CD56). METHODS: The study was on three subgroups: i) 18 subjects positive for SARS-CoV-2 swab at delivery; ii) 9 subjects that had a positive SARS-CoV-2 swab during pregnancy but resulted negative at delivery; iii) 11 control subjects with physiological pregnancy and with no previous or concomitant SARS-CoV-2 swab positivity. None of the subjects were vaccinated for SARS-CoV-2 infection. The placenta samples were analyzed for SARS-CoV-2 NP (Nucleocapsid protein) positivity and the expression of ACE2, CD147, HLA-G and CD56. RESULTS: We observed a higher percentage of SARS-CoV-2 NP positive placenta samples in the group of SARS-CoV-2 PCR positive at delivery in comparison with SARS-CoV-2 PCR negative at delivery. The localization of SARS-CoV-2 NP positivity in placenta samples was mainly in syncytiotrophoblast (ST) of SARS-CoV-2 PCR positive at delivery group and in extra-villous trophoblast (EVT) of SARS-CoV-2 PCR negative at delivery group. CD147, HLA-G positivity was higher in ST of SARS-CoV-2 PCR positive at delivery group, while CD56-expressing immune cells were decreased in comparison with control subjects. DISCUSSION: We confirmed the ability of SARS-CoV-2 to infect placenta tissues. The simultaneous SARS-CoV-2 swab positivity at delivery and the positivity of the placenta tissue for SARS-CoV-2 NP seems to create an environment that modifies the expression of specific molecules, as CD147 and HLA-G. These data suggest a possible impact of SARS-CoV-2 infection during pregnancy, that might be worthy to be monitored also in vaccinated subjects.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Female , Humans , Pregnancy , Angiotensin-Converting Enzyme 2/metabolism , HLA-G Antigens/metabolism , Placenta/metabolism , Pregnancy Complications, Infectious/metabolism , SARS-CoV-2
3.
Reprod Toxicol ; 111: 135-147, 2022 08.
Article in English | MEDLINE | ID: covidwho-1851996

ABSTRACT

Remdesivir (RDV) is the first antiviral drug to be approved by the US Food and Drug Administration for the treatment of COVID-19. While the general safety of RDV has been studied, its reproductive risk, including embryotoxicity, is largely unknown. Here, to gain insights into its embryotoxic potential, we investigated the effects of RDV on mouse preimplantation embryos cultured in vitro at the concentrations comparable to the therapeutic plasma levels. Exposure to RDV (2-8 µM) did not affect the initiation of blastocyst formation, although the maintenance of the cavity failed at 8 µM due to increased cell death. While exposure to 2-4 µM permitted the cavity maintenance, expressions of developmental regulator genes associated with the inner cell mass (ICM) lineage were significantly diminished. Adverse effects of RDV depended on the duration and timing of exposure, as treatment between the 8-cell to early blastocyst stage most sensitively affected cavity expansion, gene expressions, and cell proliferation, particularly of the ICM than the trophectoderm lineage. GS-441524, a major metabolite of RDV, did not impair blastocyst formation or cavity expansion, although it altered gene expressions in a manner differently from RDV. Additionally, RDV reduced the viability of human embryonic stem cells, which were used as a model for the human ICM lineage, more potently than GS-441524. These findings suggest that RDV is potentially embryotoxic to impair the pluripotent lineage, and will be useful for designing and interpreting further in vitro and in vivo studies on the reproductive toxicity of RDV.


Subject(s)
COVID-19 Drug Treatment , Pregnancy Complications, Infectious , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Animals , Blastocyst , Embryonic Development/genetics , Female , Mice , Pregnancy , Pregnancy Complications, Infectious/metabolism
4.
J Reprod Immunol ; 151: 103501, 2022 06.
Article in English | MEDLINE | ID: covidwho-1763858

ABSTRACT

While COVID-19 infection during pregnancy is common, fetal transmission is rare, suggesting that intrauterine mechanisms form an effective blockade against SARS-CoV-2. Key among these is the decidual immune environment of the placenta. We hypothesize that decidual leukocytes are altered by maternal SARS-CoV-2 infection in pregnancy and that this decidual immune response is shaped by the timing of infection during gestation. To address this hypothesis, we collected decidua basalis tissues at delivery from women with symptomatic COVID-19 during second (2nd Tri COVID, n = 8) or third trimester (3rd Tri COVID, n = 8) and SARS-CoV-2-negative controls (Control, n = 8). Decidual natural killer (NK) cells, macrophages and T cells were evaluated using quantitative microscopy, and pro- and anti-inflammatory cytokine mRNA expression was evaluated using quantitative reverse transcriptase PCR (qRT-PCR). When compared with the Control group, decidual tissues from 3rd Tri COVID exhibited significantly increased macrophages, NK cells and T cells, whereas 2nd Tri COVID only had significantly increased T cells. In evaluating decidual cytokine expression, we noted that IL-6, IL-8, IL-10 and TNF-α were significantly correlated with macrophage cell abundance. However, in 2nd Tri COVID tissues, there was significant downregulation of IL-6, IL-8, IL-10, and TNF-α. Taken together, these results suggest innate and adaptive immune responses are present at the maternal-fetal interface in maternal SARS-CoV-2 infections late in pregnancy, and that infections earlier in pregnancy show evidence of a resolving immune response. Further studies are warranted to characterize the full scope of intrauterine immune responses in pregnancies affected by maternal COVID-19.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Cytokines/metabolism , Decidua , Female , Humans , Immunity , Interleukin-10/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism , SARS-CoV-2 , Tumor Necrosis Factor-alpha/metabolism
5.
J Infect Dis ; 224(Supplement_6): S642-S646, 2021 Dec 08.
Article in English | MEDLINE | ID: covidwho-1559852

ABSTRACT

We previously demonstrated that the late gestation placental expression pattern of ACE2 (the primary severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2] receptor) is localized to the villous syncytiotrophoblast (ST), usually in a polarized membranous pattern at the ST base sparing the apical surface (that directly exposed to maternal blood). We found that the late gestation placental expression pattern of TMPRSS2 (the spike proteinase required for SARS-CoV-2 cellular infection), is usually absent in the trophoblast but is rarely, weakly expressed in the placental endothelium. We now show the developmental protein expression patterns of ACE2 and TMPRSS2 by immunohistochemistry throughout gestation, from the first through third trimester. We found that TMPRSS2 expression was rarely detectable in villous endothelium and very rarely detectable in the ST across gestation. We found that ACE2 expression varied during gestation with circumferential ST expression more common in early gestations and polarized expression more common in later gestation. Although this study is small, these preliminary results suggest that earlier gestation pregnancies may be more vulnerable to infection than later gestation pregnancies.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19 , Placenta/metabolism , Pregnancy Complications, Infectious/virology , SARS-CoV-2/metabolism , Serine Endopeptidases/metabolism , Adult , Angiotensin-Converting Enzyme 2/genetics , Female , Gestational Age , Humans , Placenta/virology , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/pathology , Serine Endopeptidases/genetics , Trophoblasts
6.
Placenta ; 115: 70-77, 2021 11.
Article in English | MEDLINE | ID: covidwho-1433733

ABSTRACT

Species differences are among the main reasons for the high failure rate of preclinical studies. A better awareness and understanding of these differences might help to improve the outcome of preclinical research. In reproduction, the placenta is the central organ regulating fetal exposure to a substance circulating in the maternal organism. Exact information about placental transfer can help to better estimate the toxic potential of a substance. From an evolutionary point of view, the chorioallantoic placenta is the organ with the highest anatomical diversity among species. Moreover, frequently used animal models in reproduction belong to rodents and lagomorphs, two groups that are characterized by the generation of an additional type of placenta, which is crucial for fetal development, but absent from humans: the inverted yolk sac placenta. Taken together, the translatability of placental transfer studies from laboratory animals to humans is challenging, which is supported by the fact that numerous species-dependent toxic effects are described in literature. Thus, reliable human-relevant data are frequently lacking and the toxic potential of chemicals and pharmaceuticals for humans can hardly be estimated, often resulting in recommendations that medical treatments or exposure to chemicals should be avoided for safety reasons. Although species differences of placental anatomy have been described frequently and the need for human-relevant research models has been emphasized, analyses of substances with species-dependent placental transfer have been performed only sporadically. Here, we present examples for species-specific placental transfer, including that of nanoparticles and pharmaceuticals, and discuss potential underlying mechanisms. With respect to the COVID 19-pandemic it might be of interest that some antiviral drugs are reported to feature species-specific placental transfer. Further, differences in placental structure and antibody transfer may affect placental transfer of ZIKA virus.


Subject(s)
Maternal-Fetal Exchange/physiology , Placenta/metabolism , Animals , Antiviral Agents/pharmacokinetics , Biological Transport/physiology , COVID-19/transmission , COVID-19/virology , Female , Humans , Infectious Disease Transmission, Vertical , Maternal-Fetal Exchange/drug effects , Placenta/drug effects , Pregnancy , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , SARS-CoV-2/metabolism , Species Specificity , Yolk Sac/metabolism , Yolk Sac/physiology , Zika Virus/metabolism , Zika Virus Infection/drug therapy , Zika Virus Infection/transmission , COVID-19 Drug Treatment
7.
Clin Sci (Lond) ; 135(15): 1805-1824, 2021 08 13.
Article in English | MEDLINE | ID: covidwho-1337133

ABSTRACT

In times of coronavirus disease 2019 (COVID-19), the impact of severe acute respiratory syndrome (SARS)-coronavirus (CoV)-2 infection on pregnancy is still unclear. The presence of angiotensin-converting enzyme (ACE) 2 (ACE2), the main receptor for SARS-CoV-2, in human placentas indicates that this organ can be vulnerable for viral infection during pregnancy. However, for this to happen, additional molecular processes are critical to allow viral entry in cells, its replication and disease manifestation, particularly in the placenta and/or feto-maternal circulation. Beyond the risk of vertical transmission, COVID-19 is also proposed to deplete ACE2 protein and its biological actions in the placenta. It is postulated that such effects may impair essential processes during placentation and maternal hemodynamic adaptations in COVID-19 pregnancy, features also observed in several disorders of pregnancy. This review gathers information indicating risks and protective features related to ACE2 changes in COVID-19 pregnancies. First, we describe the mechanisms of SARS-CoV-2 infection having ACE2 as a main entry door and current evidence of viral infection in the placenta. Further, we discuss the central role of ACE2 in physiological systems such as the renin-angiotensin system (RAS) and the kallikrein-kinin system (KKS), both active during placentation and hemodynamic adaptations of pregnancy. Significant knowledge gaps are also identified and should be urgently filled to better understand the fate of ACE2 in COVID-19 pregnancies and the potential associated risks. Emerging knowledge will be able to improve the early stratification of high-risk pregnancies with COVID-19 exposure as well as to guide better management and follow-up of these mothers and their children.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Placenta/virology , Pregnancy Complications, Infectious/metabolism , Receptors, Coronavirus/metabolism , SARS-CoV-2/pathogenicity , Biomarkers/metabolism , COVID-19/transmission , COVID-19/virology , Female , Humans , Infectious Disease Transmission, Vertical , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/virology , Risk Factors , Virus Internalization
8.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166218, 2021 11 01.
Article in English | MEDLINE | ID: covidwho-1323748

ABSTRACT

Throughout history, pandemics of infectious diseases caused by emerging viruses have spread worldwide. Evidence from previous outbreaks demonstrated that pregnant women are at high risk of contracting the diseases and suffering from adverse outcomes. However, while some viruses can cause major health complications for the mother and her fetus, others do not appear to affect pregnancy. Viral surface proteins bind to specific receptors on the cellular membrane of host cells and begin therewith the infection process. During pregnancy, the molecular features of these proteins may determine specific target cells in the placenta, which may explain the different outcomes. In this review, we display information on Variola, Influenza, Zika and Corona viruses focused on their surface proteins, effects on pregnancy, and possible target placental cells. This will contribute to understanding viral entry during pregnancy, as well as to develop strategies to decrease the incidence of obstetrical problems in current and future infections.


Subject(s)
Placenta/virology , Pregnancy Complications, Infectious/virology , Viral Envelope Proteins/metabolism , Virus Diseases/virology , Female , Humans , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Variola virus/metabolism , Variola virus/pathogenicity , Virus Diseases/metabolism , Zika Virus/metabolism , Zika Virus/pathogenicity
9.
Sci Rep ; 11(1): 14390, 2021 07 13.
Article in English | MEDLINE | ID: covidwho-1309469

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic affected people at all ages. Whereas pregnant women seemed to have a worse course of disease than age-matched non-pregnant women, the risk of feto-placental infection is low. Using a cohort of 66 COVID-19-positive women in late pregnancy, we correlated clinical parameters with disease severity, placental histopathology, and the expression of viral entry and Interferon-induced transmembrane (IFITM) antiviral transcripts. All newborns were negative for SARS-CoV-2. None of the demographic parameters or placental histopathological characteristics were associated with disease severity. The fetal-maternal transfer ratio for IgG against the N or S viral proteins was commonly less than one, as recently reported. We found that the expression level of placental ACE2, but not TMPRSS2 or Furin, was higher in women with severe COVID-19. Placental expression of IFITM1 and IFITM3, which have been implicated in antiviral response, was higher in participants with severe disease. We also showed that IFITM3 protein expression, which localized to early and late endosomes, was enhanced in severe COVID-19. Our data suggest an association between disease severity and placental SARS-CoV-2 processing and antiviral pathways, implying a role for these proteins in placental response to SARS-CoV-2.


Subject(s)
COVID-19/metabolism , Placenta/metabolism , SARS-CoV-2/pathogenicity , Adult , Angiotensin-Converting Enzyme 2/metabolism , Female , Furin/metabolism , Humans , Immunoglobulin G/metabolism , Infectious Disease Transmission, Vertical , Male , Nucleocapsid Proteins/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , Serine Endopeptidases/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Young Adult
10.
Clin Nutr ESPEN ; 43: 1-8, 2021 06.
Article in English | MEDLINE | ID: covidwho-1240256

ABSTRACT

BACKGROUND & AIMS: Maternal gestational infection is a well-characterized risk factor for offsprings' development of mental disorders including schizophrenia, autism, and attention deficit disorder. The inflammatory response elicited by the infection is partly directed against the placenta and fetus and is the putative pathogenic mechanism for fetal brain developmental abnormalities. Fetal brain abnormalities are generally irreversible after birth and increase risk for later mental disorders. Maternal immune activation in animals models this pathophysiology. SARS-CoV-2 produces maternal inflammatory responses during pregnancy similar to previously studied common respiratory viruses. METHOD: Choline, folic acid, Vitamin D, and n-3 polyunsaturated fatty acids are among the nutrients that have been studied as possible mitigating factors for effects of maternal infection and inflammation on fetal development. Clinical and animal studies relevant to their use in pregnant women who have been infected are reviewed. RESULTS: Higher maternal choline levels have positive effects on the development of brain function for infants of mothers who experienced viral infections in early pregnancy. No other nutrient has been studied in the context of viral inflammation. Vitamin D reduces pro-inflammatory cytokines in some, but not all, studies. Active folic acid metabolites decrease anti-inflammatory cytokines. N-3 polyunsaturated fatty acids have no effect. CONCLUSIONS: Vitamin D and folic acid are already supplemented in food additives and in prenatal vitamins. Despite recommendations by several public health agencies and medical societies, choline intake is often inadequate in early gestation when the brain is forming. A public health initiative for choline supplements during the pandemic could be helpful for women planning or already pregnant who also become exposed or infected with SARS-CoV-2.


Subject(s)
Brain , COVID-19/complications , Choline/therapeutic use , Fetal Development , Mothers , Nutritional Status , Pregnancy Complications, Infectious/virology , Animals , Brain/drug effects , COVID-19/metabolism , COVID-19/virology , Child Development/drug effects , Choline/pharmacology , Developmental Disabilities/etiology , Developmental Disabilities/prevention & control , Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/therapeutic use , Female , Fetal Development/drug effects , Fetus/drug effects , Folic Acid/pharmacology , Folic Acid/therapeutic use , Humans , Infant , Inflammation/complications , Inflammation/metabolism , Nutritional Requirements , Pandemics , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism , SARS-CoV-2 , Vitamin D/pharmacology , Vitamin D/therapeutic use
11.
Med (N Y) ; 2(5): 591-610.e10, 2021 05 14.
Article in English | MEDLINE | ID: covidwho-1220962

ABSTRACT

BACKGROUND: Pregnant women are at increased risk for severe outcomes from coronavirus disease 2019 (COVID-19), but the pathophysiology underlying this increased morbidity and its potential effect on the developing fetus is not well understood. METHODS: We assessed placental histology, ACE2 expression, and viral and immune dynamics at the term placenta in pregnant women with and without respiratory severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. FINDINGS: The majority (13 of 15) of placentas analyzed had no detectable viral RNA. ACE2 was detected by immunohistochemistry in syncytiotrophoblast cells of the normal placenta during early pregnancy but was rarely seen in healthy placentas at full term, suggesting that low ACE2 expression may protect the term placenta from viral infection. Using immortalized cell lines and primary isolated placental cells, we found that cytotrophoblasts, the trophoblast stem cells and precursors to syncytiotrophoblasts, rather than syncytiotrophoblasts or Hofbauer cells, are most vulnerable to SARS-CoV-2 infection in vitro. To better understand potential immune mechanisms shielding placental cells from infection in vivo, we performed bulk and single-cell transcriptomics analyses and found that the maternal-fetal interface of SARS-CoV-2-infected women exhibited robust immune responses, including increased activation of natural killer (NK) and T cells, increased expression of interferon-related genes, as well as markers associated with pregnancy complications such as preeclampsia. CONCLUSIONS: SARS-CoV-2 infection in late pregnancy is associated with immune activation at the maternal-fetal interface even in the absence of detectable local viral invasion. FUNDING: NIH (T32GM007205, F30HD093350, K23MH118999, R01AI157488, U01DA040588) and Fast Grant funding support from Emergent Ventures at the Mercatus Center.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Angiotensin-Converting Enzyme 2/genetics , Female , Humans , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism , SARS-CoV-2
12.
Neuroimmunomodulation ; 28(1): 1-21, 2021.
Article in English | MEDLINE | ID: covidwho-1206095

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) has devastating effects on the population worldwide. Given this scenario, the extent of the impact of the disease on more vulnerable individuals, such as pregnant women, is of great concern. Although pregnancy may be a risk factor in respiratory virus infections, there are no considerable differences regarding COVID-19 severity observed between pregnant and nonpregnant women. In these circumstances, an emergent concern is the possibility of neurodevelopmental and neuropsychiatric harm for the offspring of infected mothers. Currently, there is no stronger evidence indicating vertical transmission of SARS-CoV-2; however, the exacerbated inflammatory response observed in the disease could lead to several impairments in the offspring's brain. Furthermore, in the face of historical knowledge on possible long-term consequences for the progeny's brain after infection by viruses, we must consider that this might be another deleterious facet of COVID-19. In light of neuroimmune interactions at the maternal-fetal interface, we review here the possible harmful outcomes to the offspring brains of mothers infected by SARS-CoV-2.


Subject(s)
COVID-19/immunology , Neurodevelopmental Disorders/physiopathology , Neuroimmunomodulation/immunology , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/physiopathology , COVID-19/metabolism , COVID-19/physiopathology , Cytokine Release Syndrome/immunology , Decidua/immunology , Female , Humans , Immune Tolerance/immunology , Infectious Disease Transmission, Vertical , Neuroimmunomodulation/physiology , Placenta/immunology , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/physiopathology , SARS-CoV-2 , Umbilical Cord/immunology
13.
J Mol Histol ; 52(3): 427-435, 2021 Jun.
Article in English | MEDLINE | ID: covidwho-1192191

ABSTRACT

SARS-CoV-2, the leading cause of COVID-19 pandemic, was detected for the first time in Wuhan. In this study, we investigated the potential undesirable maternal and feto-neonatal consequences of COVID-19, and the related pathophysiological alterations in mother, neonate, and especially in the placenta as a vital organ, were reviewed. Also, the possibility of vertical transmission of virus and placental abnormalities were evaluated. The pregnant women were a vulnerable population for COVID-19, and several obstetric consequences were reported following SARS-CoV-2 infection. The higher risk of abruption, preterm labor, maternal death, stillbirth, intrauterine growth restriction, and newborns with fetal distress were adverse pregnancy and perinatal outcomes of COVID-19. Despite the ACE2 expression on placental components was confirmed, there is no agreement on the mother-child vertical transmission of this virus. Therefore, feto-neonatal consequences might be associated with placental abnormalities. The placental abnormalities are characterized by feto-maternal vascular malperfusion. Additionally, these adverse consequences lead to early termination of pregnancy in some cases, mostly via cesarean section. The pregnant women screening, coordination between healthcare personnel and neonatal unit, and infected women quarantine may decrease the risk of maternal and neonatal death after delivery.


Subject(s)
COVID-19/metabolism , Infectious Disease Transmission, Vertical , Placenta/metabolism , Pregnancy Complications, Infectious/metabolism , SARS-CoV-2/metabolism , Adult , COVID-19/pathology , Female , Humans , Infant, Newborn , Placenta/pathology , Placenta/virology , Pregnancy , Pregnancy Complications, Infectious/pathology , Pregnancy Complications, Infectious/virology
14.
Med (N Y) ; 2(5): 575-590.e5, 2021 05 14.
Article in English | MEDLINE | ID: covidwho-1179905

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection appears to increase the risk of adverse pregnancy outcomes, such as pre-eclampsia in pregnant women. The mechanism(s) by which this occurs remains unclear. METHODS: We investigated the pathophysiology of SARS-CoV-2 at maternal-fetal interface in pregnant women who tested positive for the virus using RNA in situ hybridization (viral RNA), immunohistochemistry, and hematoxylin and eosin staining. To investigate whether viral infection alters the renin angiotensin system (RAS) in placenta, which controls blood pressure, we treated human trophoblasts with recombinant spike protein or a live modified virus with a vesicular stomatitis viral backbone expressing spike protein (VSV-S). FINDINGS: Viral colonization was highest in maternal decidua, fetal trophoblasts, Hofbauer cells, and in placentas delivered prematurely. We localized SARS-CoV-2 to cells expressing angiotensin-converting enzyme 2 (ACE2) and demonstrate that infected placentas had significantly reduced ACE2. In response to both spike protein and VSV-S, cellular ACE2 decreased although angiotensin II receptor type 1 (AT1R) increased with concomitant increase in soluble fms-like tyrosine kinase-1 (sFlt1). Viral infection decreased pro-angiogenic factors, AT2R, and placental growth factor, which competitively binds to sFlt1. Sera from infected pregnant women had elevated levels of sFlt1 and angiotensin II type 1-receptor autoantibodies prior to delivery, both signatory markers of pre-eclampsia. CONCLUSIONS: SARS-CoV-2 colonizes ACE2-expressing maternal and fetal cells in the placenta. Infection in pregnant women correlates with alteration of placental RAS. As RAS regulates blood pressure, SARS-CoV-2 infection may thus increase adverse hemodynamic outcomes, such as pre-eclampsia in pregnant women. FUNDING: NIH/NICHD grants R01 HD091218 and 3R01HD091218-04S1 (RADx-UP Supplement).


Subject(s)
COVID-19 , Pre-Eclampsia , Pregnancy Complications, Infectious , Angiotensin-Converting Enzyme 2 , Female , Humans , Placenta/metabolism , Placenta Growth Factor/metabolism , Pre-Eclampsia/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism , Renin-Angiotensin System , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
15.
J Med Case Rep ; 15(1): 104, 2021 Mar 01.
Article in English | MEDLINE | ID: covidwho-1112451

ABSTRACT

BACKGROUND: Pregnancy seems to increase the risk of thrombotic thrombocytopenic purpura (TTP) relapses and make the TTP more severe in any of the pregnancy trimesters, or even during the postpartum period. CASE PRESENTATION: This study highlights details of treating a COVID-19 pregnant patient who survived. This 21-year addicted White woman was admitted at her 29th week and delivered a stillbirth. She was transferred to another hospital after showing signs of TTP, which was caused by a viral infection. CONCLUSION: This viral infection caused fever and dyspnea, and the patient was tested positive for COVID-19 infection. A chest computed tomography scan showed diffuse multiple bilateral consolidations and interlobar septal thickening. She stayed at the Intensive Care Unit for 20 days and treated with plasmapheresis. As far as we know, this is the first report of a TTP pregnant patient with COVID-19 infection.


Subject(s)
COVID-19/diagnosis , Plasmapheresis , Pregnancy Complications, Hematologic/diagnosis , Pregnancy Complications, Infectious/diagnosis , Purpura, Thrombotic Thrombocytopenic/diagnosis , Stillbirth , Acute Kidney Injury/therapy , Amphetamine-Related Disorders , Antiviral Agents/therapeutic use , COVID-19/therapy , Drug Combinations , Erythrocyte Transfusion , Female , Hemoglobins/metabolism , Humans , Hydroxychloroquine/therapeutic use , Intensive Care Units , L-Lactate Dehydrogenase/metabolism , Lopinavir/therapeutic use , Methamphetamine , Pregnancy , Pregnancy Complications, Hematologic/metabolism , Pregnancy Complications, Hematologic/therapy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/therapy , Purpura, Thrombotic Thrombocytopenic/metabolism , Purpura, Thrombotic Thrombocytopenic/therapy , Renal Dialysis , Ritonavir/therapeutic use , SARS-CoV-2 , Tomography, X-Ray Computed , Young Adult
16.
Clin Ther ; 43(2): 308-318, 2021 02.
Article in English | MEDLINE | ID: covidwho-1064961

ABSTRACT

PURPOSE: The majority of pregnancies affected by maternal coronavirus disease 2019 (COVID-19) do not result in fetal transmission. However, several studies have identified parenchymal changes in their placental tissues, suggesting a placental response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at the maternal-fetal interface. Although many COVID-19 placental studies have focused on the expression of the canonical SARS-CoV-2 entry proteins angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2, further characterization of subcellular molecules involved in viral trafficking have not yet been investigated in these tissues. Of interest are Rab proteins, a family of small GTPase proteins that direct intracellular transport between different endocytic organelles. Rab5 and Rab7 in particular have previously been implicated in HIV and cytomegalovirus invasion of placental trophoblast cells in vitro; the localization of these molecules has not been fully characterized within the human maternal-fetal interface, however, or within placental tissues from SARS-CoV-2-infected pregnancies. METHODS: Using fluorescent immunohistochemistry, Rab5 and Rab7 placental localization and comparative fluorescence intensity were explored in a cohort of placental tissues from pregnancies affected by maternal COVID-19 disease (COVID, n = 15) compared with contemporary control subjects (Control, n = 10). Fluorescence intensity was quantified by using corrected total cell fluorescence values. FINDINGS: Within placental villi, Rab5 was consistently localized in syncytiotrophoblast and cytotrophoblast cells. Rab5 had significantly higher mean (SEM) fluorescence intensity in the COVID cohort (Control, 1.96 [0.16]; COVID, 2.62 [0.09]; P = 0.0014). In contrast, although Rab7 was also localized within placental villous syncytiotrophoblast and cytotrophoblast cells, mean (SEM) Rab7 fluorescence intensity was significantly downregulated in COVID vs Control placentas (Control, 35.9 [4.1]; COVID, 20.1 [0.52]; P = 0.0001). IMPLICATIONS: This differential expression of Rab5 and Rab7 suggests that placental endocytic pathways may be altered at the maternal-fetal interface in pregnancies affected by maternal SARS-CoV-2 infection. As key molecules governing intracellular vesicle transport, including viral trafficking, Rab GTPase proteins may be of interest for ongoing studies examining placental responses to COVID-19 in pregnancy.


Subject(s)
COVID-19/metabolism , Placenta/metabolism , Pregnancy Complications, Infectious/metabolism , Trophoblasts/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Female , Humans , Pregnancy , Pregnancy Complications, Infectious/virology , SARS-CoV-2 , rab7 GTP-Binding Proteins
17.
Semin Perinatol ; 44(7): 151284, 2020 11.
Article in English | MEDLINE | ID: covidwho-1030195

ABSTRACT

The 2019 novel coronavirus disease (COVID-19) pandemic poses unique challenges to the medical community as the optimal treatment has not been determined and is often at the discretion of institutional guidelines. Pregnancy has previously been described as a high-risk state in the context of infectious diseases, given a particular susceptibility to pathogens and adverse outcomes. Although ongoing studies have provided insight on the course of this disease in the adult population, the implications of COVID-19 on pregnancy remains an understudied area. The objective of this study is to review the literature and describe clinical presentations among pregnant women afflicted with COVID-19.


Subject(s)
COVID-19/physiopathology , Pregnancy Complications, Infectious/physiopathology , Acute Kidney Injury/physiopathology , Anosmia/physiopathology , Asymptomatic Infections , Blood Coagulation Disorders/physiopathology , COVID-19/immunology , COVID-19/metabolism , COVID-19/therapy , COVID-19 Testing , Cardiomyopathies/physiopathology , Central Nervous System Diseases/physiopathology , Disease Progression , Female , HELLP Syndrome/metabolism , Humans , Hypercapnia , Hypoxia/diagnosis , Hypoxia/physiopathology , Hypoxia/therapy , Liver Diseases/metabolism , Liver Diseases/physiopathology , Mass Screening , Myalgia/physiopathology , Myocarditis/physiopathology , Oxygen Inhalation Therapy , Pre-Eclampsia/metabolism , Pregnancy , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/therapy , SARS-CoV-2 , Severity of Illness Index , Taste Disorders/physiopathology
18.
Reprod Biol ; 20(4): 568-572, 2020 Dec.
Article in English | MEDLINE | ID: covidwho-922126

ABSTRACT

SARS-CoV-2 is a new virus, to which herd immunity has not yet developed and both molecular and serological testing are not without flaws. The virus evokes a state of severe and widespread inflammation, and stimulates both innate and adaptive immune response. The angiotensin-converting enzyme 2 (ACE2), which acts as the SARS-CoV-2 receptor, is present in endothelial cells and has been noted within the human placenta. There are questions about whether pregnancy would increase the susceptibility of pregnant women to COVID-19 and disease severity within this population. In this report, we highlight physiological and immune/inflammatory considerations that may explain the susceptibility and disease pathology in response to SARS CoV-2 during pregnancy, explore testing considerations in asymptomatic individuals, discuss the potential role and of placental ACE2 receptor in the pathogenesis of COVID-19 in pregnancy and in pregnancy outcomes, and finally share our perspective with respect to an urgently needed change concerning involvement of pregnant women in research addressing COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Placenta/virology , Pregnancy Complications, Infectious/virology , Disease Susceptibility , Female , Humans , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/metabolism
19.
Placenta ; 103: 141-151, 2021 01 01.
Article in English | MEDLINE | ID: covidwho-894163

ABSTRACT

BACKGROUND: Though a large number of pregnant females have been affected by COVID-19, there is a dearth of information on the effects of SARS-CoV-2 infection on trophoblast function. We explored in silico, the potential interactions between SARS-CoV-2 proteins and proteins involved in the key functions of placenta. METHODS: Human proteins interacting with SARS-CoV-2 proteins were identified by Gordon et al. (2020). Genes that are upregulated in trophoblast sub-types and stages were obtained by gene-expression data from NCBI-GEO and by text-mining. Genes altered in pathological states like pre-eclampsia and gestational diabetes mellitus were also identified. Genes crucial in placental functions thus identified were compared to the SARS-CoV-2 interactome for overlaps. Proteins recurring across multiple study scenarios were analyzed using text mining and network analysis for their biological functions. RESULTS: The entry receptors for SARS-CoV-2 - ACE2 and TMPRSS2 are expressed in placenta. Other proteins that interact with SARS-CoV-2 like LOX, Fibulins-2 and 5, NUP98, GDF15, RBX1, CUL3, HMOX1, PLAT, MFGE8, and MRPs are vital in placental functions like trophoblast invasion and migration, syncytium formation, differentiation, and implantation. TLE3, expressed across first trimester placental tissues and cell lines, is involved in formation of placental vasculature, and is important in SARS-CoV (2003) budding and exit from the cells by COPI vesicles. CONCLUSION: SARS-CoV-2 can potentially interact with proteins having crucial roles in the placental function. Whether these potential interactions identified in silico have effects on trophoblast functions in biological settings needs to be addressed by further in vitro and clinical studies.


Subject(s)
Computational Biology , Pregnancy Proteins/metabolism , Protein Interaction Maps , SARS-CoV-2/metabolism , Trophoblasts/physiology , COVID-19/metabolism , COVID-19/pathology , Computer Simulation , Datasets as Topic , Female , HEK293 Cells , Humans , Placenta/metabolism , Placenta/physiology , Placenta/virology , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/pathology , Pregnancy Trimester, First/metabolism , Protein Binding , Proteomics/methods , Trophoblasts/metabolism , Trophoblasts/virology , Up-Regulation
20.
J Med Virol ; 93(4): 2204-2209, 2021 04.
Article in English | MEDLINE | ID: covidwho-891890

ABSTRACT

The aim is to compare VEGF-A values between pregnant women with coronavirus disease 2019 (COVID-19) and healthy controls. Furthermore, the association of inflammation parameters, disease severity, and obstetric complications with VEGF-A was investigated. This prospective case-control study was conducted on pregnant women who were admitted to Ankara City Hospital between June 14, 2020 and August 28, 2020. Pregnant women with COVID-19 (n = 95) were compared with a control group of healthy pregnant women (n = 92) with similar clinical and demographic characteristics. Demographic features, clinical characteristics, laboratory test results, VEGF-A values were compared between the groups. A correlation analysis was performed between VEGF-A levels, inflammation parameters, and clinical characteristics of the cases for pregnant women with COVID-19. VEGF-A levels were also compared between patients with composite adverse outcome and patients without any complication in the COVID-19 group. The two groups were similar except for obstetric complications (p > .05). The obstetric complication rate was higher in the COVID-19 group (p =.02). The two groups were comparable in terms of neutrophil to lymphocyte ratio and VEGF-A values. VEGF-A values were slightly different between the trimesters. A negative moderate statistically significant correlation was found between the neutrophil and VEGF-A values (r = -0.231, p =.02). VEGF-A values were similar between patients with and without composite adverse outcomes (p > .05). VEGF-A values were similar between pregnant women with COVID-19 and healthy controls.


Subject(s)
COVID-19/metabolism , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/virology , Vascular Endothelial Growth Factor A/metabolism , Adult , COVID-19/blood , COVID-19/virology , Case-Control Studies , Female , Hospitalization , Humans , Pregnancy , Pregnancy Complications, Infectious/blood , Pregnancy Outcome , Prospective Studies , Risk Factors , SARS-CoV-2/isolation & purification , Vascular Endothelial Growth Factor A/blood
SELECTION OF CITATIONS
SEARCH DETAIL